Skip to main content

Tau and neuroinflammation in Alzheimer’s disease: interplay mechanisms and clinical translation

Abstract

Alzheimer’s Disease (AD) contributes to most cases of dementia. Its prominent neuropathological features are the extracellular neuritic plaques and intercellular neurofibrillary tangles composed of aggregated β-amyloid (Aβ) and hyperphosphorylated tau protein, respectively. In the past few decades, disease-modifying therapy targeting Aβ has been the focus of AD drug development. Even though it is encouraging that two of these drugs have recently received accelerated US Food and Drug Administration approval for AD treatment, their efficacy or long-term safety is controversial. Tau has received increasing attention as a potential therapeutic target, since evidence indicates that tau pathology is more associated with cognitive dysfunction. Moreover, inflammation, especially neuroinflammation, accompanies AD pathological processes and is also linked to cognitive deficits. Accumulating evidence indicates that inflammation has a complex and tight interplay with tau pathology. Here, we review recent evidence on the interaction between tau pathology, focusing on tau post-translational modification and dissemination, and neuroinflammatory responses, including glial cell activation and inflammatory signaling pathways. Then, we summarize the latest clinical trials targeting tau and neuroinflammation. Sustained and increased inflammatory responses in glial cells and neurons are pivotal cellular drivers and regulators of the exacerbation of tau pathology, which further contributes to its worsening by aggravating inflammatory responses. Unraveling the precise mechanisms underlying the relationship between tau pathology and neuroinflammation will provide new insights into the discovery and clinical translation of therapeutic targets for AD and other tau-related diseases (tauopathies). Targeting multiple pathologies and precision therapy strategies will be the crucial direction for developing drugs for AD and other tauopathies.

Background

Dementia is currently one of the leading causes of disability and dependency among the elderly [1]. The World Health Organization reported that in 2022, more than 55 million people live with dementia worldwide, which will increase to 139 million by 2050 [2]. Dementia has become a major public health challenge and imposes enormous societal and economic burdens. Alzheimer’s disease (AD) contributes to 60–70% of dementia cases and is characterized by poor learning and memory as well as progressive and irreversible declines in cognition and behavior [3]. The two prominent pathological hallmarks of AD are the extracellular neuritic plaques (NPs) and intercellular neurofibrillary tangles (NFTs) consisting of the accumulation of β-amyloid (Aβ) and hyperphosphorylated tau protein, respectively [4]. In addition, activation of inflammatory processes and immune responses are commonly observed in AD brain tissues [5].

As an age-related neurodegenerative disease, AD is divided into two subtypes according to the age of onset: early-onset AD (EOAD, < 65 years) and late-onset AD (LOAD, ≥ 65 years). Most AD cases are LOAD, a complex disease with heterogeneous etiologies including genetics, aging, environment, lifestyle, and chronic diseases, such as obesity [6]. EOAD accounts for about 10% of the total AD cases. Only 5% of patients with EOAD carry a pathogenic variant in the AD genes (APP, PSEN1, and PSEN2 coding for the amyloid precursor protein, the presenilin 1 and 2, respectively) or the apolipoprotein E (APOE) ε4 allele. The pathogenesis remains unknown in most patients with EOAD [7].

Although the symptoms of AD are well-studied, no treatments can halt and reverse the progression of AD. Our understanding of AD pathogenesis is still limited. At present, the pathogenic hypotheses of AD mainly include the amyloid cascade hypothesis, tau hypothesis, inflammatory hypothesis, cholinergic hypothesis, etc. The amyloid cascade hypothesis is the most widely accepted one. However, nearly, all anti-Aβ drugs have failed to show satisfactory therapeutic efficacy in the past two decades. It is encouraging that in 2021, aducanumab, a humanized recombinant monoclonal antibody targeting Aβ, became the first disease-modifying therapy (DMT) drug for AD approved by an accelerated pathway of the US Food and Drug Administration (FDA). This approval based on reduced amyloid markers and its clinical efficacy remains controversial [8, 9]. In January 2023, FDA approved a new monoclonal antibody against Aβ called lecanemab for the treatment of early AD, also by its accelerated approval pathway. Lecanemab reduced brain amyloid burden markedly in early AD and cognitive decline moderately than placebo at 18 months but was related to adverse events [10]. The efficacy or long-term safety of these two drugs needs further validation. In addition, it is timely to revisit the amyloid cascade hypothesis and consider other targets, such as anti-tau or anti-inflammatory drug development for AD. Increasing evidence demonstrates that the cognitive dysfunction and severity of the disease are more related to tau pathology [11,12,13,14]. The inflammatory responses (especially neuroinflammation) accompany the entire progress of AD pathogenesis and are also linked to cognitive dysfunction [15]. Therefore, this review focuses on tau pathological changes in the progression of AD and summarizes recent studies on the mutual regulation and influence of neuroinflammation and tau pathology. The current clinical drug development based on the tau and inflammation hypotheses is also discussed. This review will provide new insights into AD pathogenesis and drug treatment strategies.

Mechanisms of tau-mediated neurodegeneration

Neuronal inclusions composed of the aberrant aggregated microtubule-associated protein tau (MAPT) have been found in the brains of patients with neurodegenerative disorders called tauopathies, including AD, progressive supranuclear palsy (PSP), frontotemporal lobar degeneration (FTLD), and Pick’s disease [PiD, also termed frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17)]. Misfolded tau is a key pathological feature in AD, the most common tauopathy. The insoluble tau deposits comprised of fibrils are most commonly found in the cell bodies and dendrites of neurons, and they are called NFTs [16]. Correlations between NFT density and clinical symptoms, such as a cognitive decline in AD, have been demonstrated [11, 12].

Expression and function of tau

Tau was first discovered in 1975. As a microtubule-associated protein, it is expressed at a high and soluble level in neurons throughout the central nervous system (CNS) [17]. Tau is predominantly found in the axons of neurons. A pivotal function of tau is to bind to microtubules, enhance the assembly, and regulate the stability of microtubules, which plays essential roles in neurite outgrowth, cell shape and polarity, and intracellular cargo (such as neurotransmitters) transport [18].

The human tau is encoded by the 16 exons-comprising MAPT gene on chromosome 17q21 [19]. In the human brain, alternative splicing of exons 2 and 3 of the tau gene produces three isoforms with 0, 1, or 2 N-terminal repeats (0N, 1N, 2N), whereas the absence or presence of exon 10 results in tau species with either three (3R) or four (4R) carboxyl-terminal microtubule-binding domain. Thus, six major tau isoforms are expressed in the human brain and range from 352 to 441 amino acids in length [19]. The expression of Tau isoforms is regulated developmentally. In the normal adult brain, all six isoforms are present with approximately equimolar 4R and 3R isoforms, whereas, in the human fetal brain, only 0N3R tau is expressed [18]. The 4R tau isoforms exhibit higher affinity when binding to microtubules than the 3R isoforms [20]. Studies have shown that some known mutations in the tau gene affect the alternative splicing of exon 10, resulting in an altered 4R:3R ratio, a crucial feature of primary tauopathies [21, 22]. Primary tauopathies are a subgroup of FTLD disorders characterized by neuronal and glial tau inclusions with predominant frontal and temporal lobe atrophy. According to the fibrillated tau isoform (3R or 4R), primary tauopathies can be further classified into three major subtypes, including 3R tauopathies (such as PiD), 4R tauopathies (including PSP and corticobasal degeneration (CBD)), and mixed 3R/4R tauopathies [23]. AD is considered a secondary tauopathy due to tau pathology may occur as a consequence of extracellular amyloid plaques. In AD brains, tau aggregates into NFTs or neuropil threads composed of 3R and 4R Tau [24]. In this review, we focused on AD, the representative of secondary tauopathies.

Insoluble fibril formation of tau has long been considered an essential toxic event in AD. However, numerous studies have shown that the smaller, soluble, and non-fibrillar tau oligomers, called the “tau we cannot see” [25], play a more critical role in the neurotoxicity and propagation of tau damage in the CNS [25,26,27,28,29]. Mutations in the MAPT gene lead to FTDP-17 [22], providing evidence that tau dysfunction due to tau mutations induces neurodegeneration. Recent studies have found that overexpression of either wild-type tau or human P301L-mutant tau inhibits neural network activity independent of fibril formation. Turning off their overexpression attenuates the inhibition of network activity, which is associated with soluble tau but not fibrillar tau [30]. Furthermore, inhibition of endogenous tau improves behaviors and protects neurons from toxicity in APP/PS1 mice, a mouse model with AD-like Aβ pathology [31]. These findings indicate that soluble oligomeric tau may play a more essential role in neurodegeneration than insoluble fibrillar tau (including NFT), the “tau we can see”.

Post-transcriptional modifications of tau

In humans, tau protein undergoes several post-translational modifications to regulate the interactions with microtubules, including phosphorylation, N-linked glycosylation (N-glycosylation), O-linked N-acetylglucosaminylation (O-GlcNAcylation), glycation, ubiquitination, truncation, nitration, and oxidation [19]. Phosphorylation is the most widely studied post-translational modification for tau, as more than eighty serine and threonine residues and five tyrosine residues are potential phosphorylation sites on the longest isoform of human tau [32]. The normal phosphorylation state of tau is critical for neuronal plasticity.

However, under pathological conditions, various highly increased post-translational modifications, such as hyperphosphorylation, destabilize the interaction of tau with microtubules [33] and enhance the capacity of tau to accumulate in the cytoplasm [34], leading to microtubule instability and transport dysfunction. In the normal adult brain, there are 2–3 mol of phosphate per mole of tau, but in the AD brain, tau protein is twofold to threefold hyperphosphorylated [35]. Studies have shown that individual missense mutations in tau alter potential phosphorylation sites and promote phosphorylation levels compared to unmutated tau [36]. In addition, various kinases and phosphatases have been found to regulate tau phosphorylation, such as glycogen synthase kinase-3β (GSK3β), cyclin-dependent kinase-5 (CDK5), p38 mitogen-associated protein kinase alpha (p38α MAPK), extracellular signal-related kinase (ERK), c-Jun N-terminal kinase (JNK), protein kinase A (PKA), and protein phosphatase 2 (PP2A) [4, 37,38,39]. A recent study demonstrated that tau phosphorylation is controlled by interdependence, an initial site-specific phosphorylation (they called “master sites”) leads to subsequent multi-site phosphorylation. Co-targeting p38α, the most central tau kinase associated with interdependence, and the master sites synergistically eliminated hyperphosphorylation of tau [39]. Hyperphosphorylated tau leads to abnormal aggregation of tau protein, which loses its ability to stabilize microtubules, thereby impairing neuronal function [23]. In addition, tau aggregates have been shown to have prion-like seeding and spreading properties [40]. The pathogenic tau can be released from diseased neurons, then uptake by previously unaffected normal neurons, inducing pathogenic tau production in normal neurons. This property of pathogenic tau leads to disease progression and broader clinical symptoms [41, 42]. Further discussion of tau propagation is provided in the following section.

In addition to abnormal hyperphosphorylation, other types of post-translational modifications of tau may also contribute to tau dysfunction in disease states. For example, reduced tau O-glycosylation could lead to increased phosphorylation, while the enhancement of O-glycosylation reduces the extent of tau phosphorylation [43,44,45]. In addition, the acetylation of tau is an early pathological feature of neurodegeneration. Acetylated tau inhibits its degradation, promotes pathological aggregation and propagation, and contributes to tauopathy [46,47,48,49,50,51]. Other post-translational modifications, such as isomerization and truncation, have been shown to promote and stabilize paired helical filaments (PHFs) [52, 53]. The methylation of tau could suppress the aggregation of tau [54]. The precise mechanism of these post-translational modifications of tau in Alzheimer's neurofibrillary degeneration is unclear. However, the hyperphosphorylation alone can induce pathological functional changes in tau, promoting self-aggregation into PHF tangles. Moreover, tau hyperphosphorylation is present in almost every tauopathy, suggesting that different post-translational modifications may be involved in modulating hyperphosphorylation [4]. Therefore, targeting hyperphosphorylation of tau, which may be a convergent pathway of tauopathies, including AD, is a crucial direction for drug development.

Propagation of tau pathology

Studies propose that AD and other non-infectious neurodegenerative disorders associated with aggregation of fibrillar proteins exhibit features similar to prion disease [55]. The transfer of abnormal misfolded proteins, including tau and Aβ, has a common feature of the pathological propagation between cells [42, 56]. Postmortem studies have shown that the spread of tau pathology in AD follows a predictable pattern, allowing neuropathological diagnosis of different AD stages defined by Braak staging (I–VI). Initial neurofibrillary tangles and neuropil threads develop in the entorhinal cortex, then the hippocampus, and gradually affect additional brain regions as the disease progress, eventually affecting the neocortex [57]. An in vivo study has demonstrated that the intracerebral injection of synthetic tau fibrils into the hippocampus or frontal cortex of tau-P301L transgenic mice at 3 months increases tau hyperphosphorylation and accumulation around the injection site. In addition, the spread of tau pathology is time-dependent from the injection site to distant interconnected brain regions [58].

Although the specific mechanisms underlying the interneuronal spread of these tau aggregates remain poorly understood, there is a large amount of evidence indicating that the abnormally hyperphosphorylated or oligomeric tau can be secreted from neurons into the extracellular space, then be taken up by other normal neurons, and finally causes interneuronal transfer of tau pathology and spreading of tau toxicity across different brain regions [59,60,61] (Fig. 1). The protopathic tau seeds may be released and internalized by neurons through trans-synaptic and non-synaptic pathways in parallel to promote tau spreading [62,63,64,65]. Sokolow et al. revealed that C-terminal truncated tau is abundant in the cortical pre-synaptic terminals, and tau cleavage promotes tau aggregation, secretion, and propagation in AD [66]. In addition, trans-synaptic tau propagation and aggregation can also be independent of the presence of endogenous soluble tau, but the absence of endogenous tau reduces its neurotoxicity [63].

Fig. 1
figure 1

Schematic diagram showing the interaction between neuroinflammation and tau pathology contributing to the progress of AD pathogenesis. β-amyloid, ApoE4 apolipoprotein E4, cGAS cyclic GMP–AMP synthase, CMKLR1 chemerin chemokine-like receptor 1, CX3CL1 chemokine (C–X3–C motif) ligand 1. CX3CR1 CX3C motif chemokine receptor 1, GSK3β glycogen synthase kinase-3 beta, IL-1β interleukin-1β, IL-1R interleukin-1 receptor, HSPGs heparan sulfate proteoglycans, LRP1 low-density lipoprotein receptor-related protein 1, MAPK mitogen-activated protein kinase, NF-κB nuclear factor kappa B, NFTs neurofibrillary tangles, NLRP3 NLR family pyrin domain-containing protein 3, Nox2 NADPH oxidase 2, P2RX7 P2X purinoceptor 7, PHFs paired helical filaments, PQBP1 polyglutamine-binding protein 1, pTau phosphorylated tau, STAT1 signal transducer and activator of transcription 1, STING stimulator of interferon genes, TFEB transcription factor EB, TNF-α tumor necrosis factor α, TREM2 triggering receptor expressed on myeloid cells 2

It has been proposed that intracellular tau can be released from neurons by exocytosis, including secretion (free protein), extracellular vesicles (such as exosomes and ectosomes/microvesicles), and neuronal death pathways [67]. Meanwhile, the extracellular tau can be taken up by neighboring cells, both neurons and glial cells, through various pathways, including phagocytosis [68], macropinocytosis [69], receptor-mediated uptake [67, 70], endocytosis [71], and/or membrane fusion of exosomes [72, 73]. A recent study showed that the low-density lipoprotein receptor-related protein 1 (LRP1) expressed in neurons regulates the endocytosis of tau and its subsequent spread [74]. Knockdown or inhibition of LRP1 significantly reduces tau uptake and spread in vitro and in vivo [74]. In addition, exosomes are involved in the dissemination of tau pathology. Exosomes isolated from the brain, cerebrospinal fluid (CSF), and plasma of AD patients and/or AD animal models contain pathologic Aβ and tau [73, 75,76,77]. Increased neuronal activity enhances the release of tau-containing exosomes [75] and exacerbates tau propagation and pathology [78]. Exosomes carrying tau are taken up by local and remote cells and contribute to apoptosis and neuronal loss [79]. Peeraer and colleagues found that the tau pathology as a consequence of injection with tau-preformed fibrils into the hippocampus of tau-P301L transgenic mice induced selective neuron loss of the CA1 region [58]. Inhibition of exosome synthesis using the neutral sphingomyelinase-2 inhibitor reduces tau propagation from the entorhinal cortex to the dentate gyrus in adeno-associated virus (AAV)-based and P301S tauopathy mouse models [80]. These findings reveal a pivotal role in the cell-to-cell spreading of abnormal tau in neurotoxicity and provide potential therapeutic strategies for tau-targeted immunotherapies in AD and other tauopathies. Furthermore, tau propagation-induced tau pathology is based on the spread of tau between neurons and the dissemination between neurons and glial cells [81, 82]. Glial cells such as microglia and astrocytes perform normal immune functions in CNS and also play a vital role in the spreading of pathological tau [80, 83]. This part will be discussed in detail in the next section.

Neuroinflammation: a link between tau and AD

Over the last decade, evidence indicates that CNS inflammation (neuroinflammation) may play a pivotal role in the pathological progression of AD. The chronic, sustained inflammatory response in the brain is considered the third core pathological feature of AD. It provides a link between the other two core pathologies, Aβ plaques and NFTs [84]. The acute neuroinflammatory response is a fundamental protective immune response against noxious and irritable stimuli, such as infection, toxins, and injury, which is a well-established defense response and is essential for the brain's repair process. However, when the balance of pro-inflammatory and anti-inflammatory signaling is disrupted, it leads to a chronic inflammation response. This chronic neuroinflammation is caused by persistent activation of glial cells and excessive release of cytotoxic molecules, which adversely affects brain function and is a major cofactor in the pathogenesis of many neurodegenerative disorders, including AD [84].

The neuroinflammatory process in AD is mainly driven by the innate immune cells in the brain, including microglia and astrocytes [85]. Microglia are the resident immune cells of the brain and play a pivotal role in the immune defense of CNS. They are in an inactive “resting” state under physiological conditions while actively monitoring the brain environment and brain parenchyma with highly motile processes [86]. Microglia will shift from the resting status to an activated state when they recognize a stimulus in CNS, characterized by the morphological changes and modulations in the gene expression, including pro- and anti-inflammatory molecules and microglial surface receptors. These receptors include the triggering receptor expressed on myeloid cells 2 (TREM2), toll-like receptors (TLRs), and G protein-coupled receptors (GPCRs), such as chemokine CX3C motif receptor 1 (CX3CR1), formyl peptide receptor 2 (FPR2) and chemokine-like receptor 1 (CMKLR1), which upon activated by the stimuli, mediate microglial activation and polarization phenotype [87, 88]. Microglial activation is believed to be a double-edged sword in AD pathology. In the early stages of AD, activated microglia cause phagocytosis and clearance of pathologic Aβ and/or tau, positively affecting AD pathologies in animal models [89, 90]. However, sustained activation of microglia leads to the continuous release of inflammatory factors and reduces their ability to phagocytose and degrade neurotoxins, which in turn exacerbates Aβ accumulation, tau propagation, and neuronal death, ultimately promoting AD progression [5, 91, 92].

Recent evidence suggests that reactive astrocytes also contribute to neuroinflammatory processes associated with AD pathology [93]. Astrocytes are the most abundant glial cells in the brain and have a variety of complex and essential functions in CNS, including maintenance of brain homeostasis, synaptic transmission, and information processing through neural circuits [94]. Astrocytes become activated in AD and exhibit certain immune functions. Like microglia, astrocytes can be triggered by various factors, such as pathological Aβ, tau species, and proinflammatory cytokines [95, 96]. The activated microglia and reactive astrocytes produce nitric oxide (NO) and inflammatory cytokines, such as interleukin-1 (IL-1), IL-6, tumor necrosis factor α (TNF-α), and transforming growth factor β (TGF-β), that contribute to a reinforced inflammatory cascade [85, 97]. Studies have shown that these pro-inflammatory cytokines are markedly elevated in the brain and CSF of AD patients [98,99,100]. Interestingly, the upregulation of such proinflammatory cytokine has been observed even before signs of increased Aβ and hyperphosphorylated tau in CSF of mild cognitive impairment (MCI) patients [101], suggesting that the inflammatory processes had occurred in the early stages of AD.

Although the link between neuroinflammation and AD was discovered decades ago [102], it remains unclear whether it is a cause or a consequence of the disease. Recent studies have shown that microglial activation occurs in the preclinical AD stage. In addition, with the progression of the disease, immune activation, including the activation of microglia and astrocytes, diverts to a more harmful stage [103]. This indicates that neuroinflammation may be involved in the etiology of AD. Furthermore, since the immune response exists throughout the pathological progress of AD [104, 105], suggesting that it may also participate in and aggravate the disease development. In addition, not limited to neuroinflammatory response, systemic inflammation is currently beginning to be considered a contributor to AD development of AD [106, 107]. Multiple epidemiological studies show that anti-inflammatory drugs such as non-steroidal anti-inflammatory drugs (NSAIDs) have a sparing effect on AD [108,109,110]. Unexpectedly, clinical trials targeting inflammation with NSAIDs have not improved cognition in AD patients [111, 112]. The outcomes of clinical trials may be related to the disease stages of recruited AD patients. Moreover, these results also indicate that further elucidation of the exact relationship and mechanism between brain inflammatory events and the pathological development of AD is necessary to explore successful therapies and drugs.

Evidence suggests that the pathological activation of glial cells and the release of inflammatory factors involved in neuroinflammation can exacerbate tau pathology via direct or indirect pathways, leading to neuronal damage and cognitive impairment, ultimately aggravating AD pathology [113, 114]. The neuroinflammation exacerbating tau pathology may be correlated with the regulation of tau post-translational modification and propagation [115]. Modifying or intervening in the immune response to slow down or inhibit tau pathology provides a potential direction for developing potential therapeutic drugs for AD. In the succeeding sections, we discuss several possible mechanisms for the link between altered neuroinflammation and tau pathology observed in AD (Fig. 1). We highlight the potential value of targeting the combination of neuroinflammation and tau pathology and/or their link in AD treatment.

Role of neuroinflammation in post-transcriptional modifications of tau

Accumulating evidence suggests that microglia activation participates in the progression of tau-related neuropathology. Felsky et al. reported that the proportion of morphologically activated microglia (PAM) in postmortem cortical tissue from AD patients is strongly related to tau pathology, and their mediation models support microglial activation as an upstream event in AD leading to accumulation of hyperphosphorylated tau and subsequent cognitive decline [116]. Our and other studies found that systemic administration of lipopolysaccharide (LPS, inducer of inflammation) leads to microglial activation in the mouse brain, which results in tau hyperphosphorylation at specific sites [117, 118]. The effect of microglial activation on tau hyperphosphorylation is related to cell surface receptors which mediate inflammatory responses.

TREM2, a pivotal risk factor for LOAD [119, 120], is associated with tau pathology. TREM2 is a receptor for Aβ [121] and is exclusively expressed by microglia in the brain of mice and humans [122, 123]. In the CSF of AD patients, the R47H (rs75932628) variant of TREM2 or soluble TREM2 has been found to correlate with total or phosphorylated tau (Thr181), respectively, but not with Aβ42 [124, 125]. Numerous studies have been conducted in animal models to explore how TREM2 affects tau pathology. TREM2 deficiency in a hTau (expressing human MAPT but not endogenous mouse Mapt) mouse model exacerbates tau phosphorylation and aggregation at the early disease stage [126]. In contrast, in tau-P301S transgenic mice, TREM2 deletion attenuates neuroinflammation and protects against neurodegeneration at a late stage without altering tau phosphorylation and aggregation [127]. Further research reveals that only in the presence of Aβ pathology, TREM2 deletion further exacerbates tau accumulation and brain atrophy [128]. TREM2 may play a pivotal role in all stages of AD pathogenesis, and maintaining the normal function of TREM2 may point out a direction for AD treatment.

CX3CR1, another receptor explicitly expressed on microglia, is involved in tau pathology. CX3CR1 belongs to G protein-coupled receptors (GPCRs). GPCRs, as one of the most prominent protein families, is a class of receptors with seven-transmembrane domains. GPCRs sense extracellular molecules and then transduce the signals to intracellular effector molecules, resulting in cellular responses [129]. A body of evidence indicates the opposing effects of CX3CR1 with its ligand fractalkine (CX3CL1) on Aβ and tau pathologies. Deletion of CX3CR1 or depression of the CX3CL1/CX3CR1 axis reduces Aβ deposition [130,131,132] but exacerbates tau pathology, such as increased phosphorylation and aggregation of tau, and this is associated with worsened behavioral and cognitive impairments [132,133,134,135,136,137] (Fig. 1). These findings suggest that regulating the CX3CL1/CX3CR1 axis may be a potential target for preventing tau-related neurodegeneration.

In addition, astrocytes are also suggested to play a role in the hyperphosphorylation of tau. Astrocytes exacerbate Aβ-induced tau hyperphosphorylation and truncation. The mechanism relates to the increased caspase-3 activity caused by soluble inflammatory factors released by active astrocytes [96]. Our recent study shows that p47phox, the organizer subunit of NOX2 (Nicotinamide adenine dinucleotide phosphate oxidase 2, NADPH oxidase 2), is associated with cognitive function and tau pathology in AD. The expression of p47phox in neurons contributes to tau hyperphosphorylation directly, while p47phox in astrocytes affects tau hyperphosphorylation by activating astrocytes indirectly [13]. ApoE4, the most potent risk factor for the pathogenesis of LOAD, modulates neuroinflammatory response and glial activation [138]. Astrocyte- or neuron-specific ApoE4 could regulate tau phosphorylation in glia-dependent or independent manner [139,140,141]. Saroja et al. demonstrated that astrocyte-secreted ApoE4 and glypican-4 (GPC-4) bind to LRP1 in neurons, leading to tau accumulation and propagation [141] (Fig. 1). Deletion of astrocytic ApoE4 markedly reduces phosphorylated tau [142]. ApoE4 affects neuroinflammation, tau pathology, and tau-mediated neurodegeneration independently of Aβ pathology [143]. These findings indicate that astrocytes are crucial in exacerbating tau hyperphosphorylation and ultimately promoting AD pathology. In addition, tau is also present in the astrocytes of individuals with AD [144, 145]. However, how tau pathology is induced and regulated in astrocytes in AD and other tauopathies remains unknown.

FPR2 and CMKLR1, two GPCRs expressed on astrocytes and/or microglia, are also shown to be related to AD pathology. They are known initially as orphan receptors and recognize various endogenous and exogenous chemotactic ligands to exert pro-inflammatory or anti-inflammatory functions [146, 147]. Aβ is one of their ligands [148, 149]. Recently, the structure of the FPR2-Gi protein complexed with Aβ has just been solved [150]. Deficiency of FPR2 or administration of its inhibitors/anti-inflammatory ligands could alleviate the pathological symptoms of AD, including reduced activation of glial cells (microglia and/or astrocytes) and tau hyperphosphorylation, and ultimately leading to improvement in cognitive function [151,152,153,154]. Our recent study identified that CMKLR1 deletion increases Aβ plaques in the AD mouse brain but reduces mortality and cognitive deficits of AD mice and attenuates tau hyperphosphorylation [14]. Further studies found that CMKLR1 and its ligand chemerin regulate the migration and recruitment of microglia to Aβ plaques in vivo and in vitro [155]. Pro-resolving ligands or inhibitors of CMKLR1 have been shown to attenuate inflammatory responses, including neuroinflammation [156,157,158]. Whether FPR2 and CMKLR1 regulate the abnormal phosphorylation of tau by mediating microglial activation needs further verification.

The effect of microglia and astrocytes on tau pathology is related to the production and release of pro-inflammatory cytokines after their activation. A recent study shows that NLR family pyrin domain-containing protein 3 (NLRP3) inflammasome activation induces tau hyperphosphorylation and aggregation through modulating tau kinases and phosphatases [159]. NLRP3 inflammasome has been proven to accumulate inside microglia upon activation, promoting cleavage and activity of caspase-1 and release of IL-1β [160]. IL-1β treatment or overexpression exacerbates tau phosphorylation through the activation of p38 mitogen-activated protein kinase (MAPK) and/or glycogen synthase kinase-3β (GSK-3β) in neuron–microglia co-cultures [161] and AD model mice [162, 163]. Bhaskar et al. reported that microglia activation induces tau hyperphosphorylation via the IL-1β/p38α MAPK pathway in vitro and in vivo [135, 137]. Selective suppression of p38α MAPK significantly reduces tau hyperphosphorylation and improves working memory in hTau mice [164]. Our previous studies indicate that the deficiency of serum amyloid A (SAA), an acute-phase protein with cytokine-like properties, enhances tau phosphorylation induced by systemic LPS administration [117]. Overexpression of SAA by intracerebral injection attenuates tau hyperphosphorylation, and the mechanism is related to SAA-induced secretion of IL-10 from microglia [117]. Another study demonstrates that IL-10 deletion activates microglia, increases IL-6 production, and leads to tau hyperphosphorylation in response to acute systemic inflammation [165]. Other inflammatory factors such as IL-3, IL-6, IL-18, tumor necrosis factor-α (TNFα), and macrophage migration inhibitory factor (MIF) are also found to be involved in tau phosphorylation and/or truncation [166,167,168,169,170]. All these findings suggest that glia-neuron signaling contributes to the pathogenesis of tauopathy, and in-depth exploration of this signaling and regulation may provide valuable strategies for AD treatment.

In addition, tau also undergoes both N-linked glycosylation (N-glycosylation) [171] and O-linked N-acetylglucosaminylation (O-GlcNAcylation) [172], which have been proposed to affect tau phosphorylation and aggregation [173]. As discussed above, a reciprocal relationship has been found between O-GlcNAcylation and phosphorylation on tau [43]. Increases in tau O-GlcNAcylation inhibit tau aggregates and neuronal cell loss [174], indicating that O-GlcNAcylation modification can protect AD progression. O-GlcNAcylation levels are reduced in the cortex and hippocampus of AD individuals and in vivo and in vitro models of AD [175]. The reduced O-GlcNAcylation is related to mitochondrial dysfunction and neurodegeneration [175], which may be associated with neuroinflammation. Interestingly, the R47H variant of TREM2, which is expressed by microglia and linked to innate immunity, has been found to present an altered glycosylation pattern and decreased stability compared with wild-type TREM2 [176], indicating a potential link between microglial neuroinflammation and glycosylation [177, 178]. However, the direct correlation between neuroinflammation and glycosylation and how it regulates tau post-translational modification and tau pathology remains to be explored.

Abnormal acetylation of tau on lysine residues spans the microtubule-binding repeat region (MTBR), and this modification alone is sufficient to induce tau pathology and neurodegeneration [51]. Tau is acetylated by the lysine acetyltransferase p300 and its close homolog CREB-binding protein (CBP) [48]. The expression and activity of p300 are increased in the brain of AD patients [179], and the dysregulation of p300/CBP promotes tau acetylation, which could aggravate tau accumulation and pathology [46, 48]. Inhibiting p300 with salsalate, a non-steroidal anti-inflammatory drug, could induce tau deacetylation, preserve tau axonal localization, and protect mice from neurodegeneration [51]. The histone deacetylase 6 (HDAC6) has also been implicated in tau deacetylation/acetylation [180]. Cohen et al. reported that tau is a substrate of HDAC6, and the inhibition of HDAC6 increases tau acetylation [47]. HDAC6 also inhibits tau hyperphosphorylation within the MTBR [181]. However, other studies found that under neuroinflammatory stress, deletion or inhibition of HDAC6 suppresses mislocalization and neuritic aggregation of tau through a matrix metalloproteinase (MMP-9)-mediated mechanism [182]. HDAC6 inhibitors are being developed to treat immune and inflammatory diseases, including human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) [183]. HDAC6 knockdown attenuates reactive oxygen species (ROS) generation, NADPH oxidase activation, and neuroinflammation response in HIV-1 transactivator of transcription (Tat)-stimulated astrocytes [184]. In addition, Nox2 knockdown suppresses HIV-1 Tat-induced HDAC6 expression and subsequent upregulation of pro-inflammatory chemokines [184], indicating a possible link between neuroinflammation and mediation of HDAC6. All these findings suggest that neuroinflammation-involved regulation of tau acetylation may be a potential therapeutic strategy to ameliorate tau pathology-involved neurodegeneration.

Role of neuroinflammation in the propagation of tau

A large number of studies have explored the mechanisms of tau transmission between neurons and its impact on tau pathology. Here, we review the interplay between neuroinflammation and tau transmission. Emerging studies have indicated a possible interaction between the prion-like features of tau protein and the neuroinflammatory response in tau transmission and pathology [185, 186], although their causal relationship is currently uncertain. Microglia have been strongly implicated as a pivotal player and play a complex role in the propagation of tau pathology. In the brain of AD patients, microglial activation and tau accumulation propagate spatially in parallel, following brain circuits and staging of tau pathology [187]. Microglia can phagocytose and degrade pathologic tau, neuronal synapses, or whole live neurons, although not very efficiently [188, 189]. In addition, sustained reactive or senescent microglia become hypofunctional and release seed-competent tau, leading to the exacerbated spread of tau pathology [190].

Microglia sense pathological tau species via various surface receptors to trigger phagocytosis and/or degradation. Tau can bind to microglial CX3CR1 and initiate the internalization and degradation of tau [191]. CX3CL1 competes with tau for binding to CX3CR1, leading to a decrease in the internalization of tau. In addition, phosphorylated tau at Ser396 exhibits reduced binding affinity to CX3CR1 [191]. These findings indicate that CX3CL1/CX3CR1 axis plays a crucial role in tau phagocytosis and degradation by microglia. In addition, several studies have shown that tau seeds taken up by microglia can activate microglia, further exacerbating tau propagation and pathology [186, 192, 193]. Jin et al. reported that microglia uptake exogenous monomeric tau in parallel via two surface receptors, LRP1 and TREM2, and then induce nuclear factor NF-κB activation in microglia through two different pathways, LRP1/polyglutamine binding protein 1 (PQBP1)/cyclic GMP–AMP synthase (cGAS)/Stimulator of interferon genes (STING) and TREM2/extracellular signal-regulated kinase (ERK)/Phosphoinositide 3-kinase (PI3K) pathways, respectively (Fig. 1), ultimately triggering neuronal death [192]. Microglial NF-κB pathway activated by tau can exacerbate the processing and release of pathological tau with seeding activity. In contrast, deficiency or inhibition of NF-κB reduces the seeding and spread of tau inclusions in tauopathy mice [193]. However, another study revealed that TREM2 knockout or its R47H variant decreases microgliosis around Aβ plaques and enhances the propagation of tau aggregates [70]. This may be related to the different types of microglia [194]. In addition, the microglial NLRP3 inflammasome is also involved in the pathological propagation of tau. The aggregated tau seeds activate NLRP3/apoptosis-associated speck-like protein containing a CARD (ASC) inflammasome in microglia, following microglial uptake and lysosomal sorting of tau seeds [186]. Administration of NLRP3 inhibitor or ASC deficiency reduces the exogenously or non-exogenously seeded tau pathology, highlighting the promotion of NLRP3/ASC axis activation on the propagation of tau seeds [186]. In addition, exosomes are involved in the microglia-mediated pathological propagation of tau. Asai et al. demonstrated that depleting microglia or inhibiting microglial exosome synthesis reduces tau propagation from the entorhinal cortex to the dentate gyrus in the adeno-associated virus (AAV)/tau-injection P301S tauopathy mice [80] (Fig. 1). Suppression of P2X purinoceptor 7 (P2RX7)-induced exosome secretion from microglia attenuates misfolded tau aggregates in the hippocampus and improves cognitive deficits of P301S mice [195]. Furthermore, Zhu et al. reveal that TREM2 deficiency exacerbates pathological tau propagation via microglial exosomes [196]. These findings suggest that elucidating the mechanisms by which microglia participate in tau propagation may provide new strategies for the intervention of tau pathology.

In addition to microglia, astrocytes contribute to the spreading of tau pathology. It has been shown that astrocytic tau pathology occurs in AD and other tauopathies [197]. The aberrant tau in astrocytes may also come from the phagocytosis of neuronal debris and dystrophic synapses containing tau aggregates [198, 199]. TFEB induces astrocytic trafficking of tau fibrils via macropinocytosis, possibly through interaction with heparan sulfate proteoglycans (HSPGs) [200, 201]. Overexpression of transcription factor EB (TFEB, a regulator of lysosomal biogenesis) in astrocytes promotes tau fibril species uptake and lysosomal activity as well as attenuates tau spreading and pathology in the hippocampus of P301S tauopathy mice [201] (Fig. 1). Astrocytes can internalize tau monomers or fibrils through mechanisms independent of HSPGs [202]. The integrin αV/β1 receptor interacts with tau monomers or fibrils and mediates tau uptake in primary astrocytes. The binding of tau fibrils to astrocyte αV/β1 activates integrin signaling, resulting in NF-κB activation, leading to an increase of pro-inflammatory cytokines and chemokines, and induction of expression of neurotoxic astrocytic markers [203]. These findings suggest that phagocytized tau by astrocytes can be degraded and/or induce astrocytic activation. Whether astrocytic tau is secreted to the outside of cells as exosome remains controversial [80, 204]. As discussed above, we indicated that CMKLR1 expressed on neurons affects tau phosphorylation via mediating tau seeding [14] (Fig. 1), and how CMKLR1 on glial cells contributes to tau propagation needs investigation. In addition, it has been found that astrocyte-derived exosomes drive neurodegeneration of AD through the acceleration of Aβ aggregation in vivo [205]. Whether astrocytes can promote tau pathology via secreting exosomes still requires clinical evidence and experiment verification.

Thus, these results suggest that the interaction of pathological tau with glial cells promotes the propagation of pathological tau and AD progression. Modulating the activity of glial cells, the expression of surface receptors, the activation of inflammation-related signaling pathways, or the release of exosomes to interfere with their promotion of tau spread may provide potential strategies for AD treatment.

Advances in AD drug development focused on tau pathology and neuroinflammation

Current pharmacologic treatments for AD are three cholinesterase inhibitors (donepezil, rivastigmine, and galantamine), one N-methyl-D-aspartate (NMDA) receptor antagonist (memantine), and two DMT anti-Aβ antibody drugs (aducanumab and lecanemab). These cholinesterase inhibitors and NMDA receptor antagonist memantine only temporarily relieve the symptoms of AD patients but do not delay the progression of the disease [206, 207]. For the past two decades, the therapeutic approaches have focused on developing DMT drugs, particularly those targeting Aβ. However, with the failures or unsatisfactory efficacy of drugs anti-Aβ [9, 208,209,210,211], the development of drugs targeting other targets is increasing, such as anti-tau pathology and anti-neuroinflammation [212]. According to the report by Cummings et al., among the 143 drugs in trials in the AD drug development pipeline as of January 2022, DMT drugs account for 83.2% [212]. There are 20 (16.8%) agents targeting Aβ, 13 (10.9%) targeting tau, 28 (19.3%) targeting inflammation, and 19 (16%) targeting synaptic plasticity or neuroprotection.

Tau pathology as a therapeutic target

There are no drugs yet approved to treat tauopathies. Currently, tau-targeted disease-modifying therapies developed for AD or other tauopathies mainly include mediators of tau post-translational modifications, anti-tau immunotherapy (active and passive), tau aggregation inhibitors, microtubule stabilizers, and gene therapy. We will primarily review drugs in clinical trials targeting tau post-translational modifications and anti-tau immunotherapy (Table 1). Anti-tau immunotherapy aims to target tau phosphorylation, aggregation, and propagation, and clear extracellular and/or intracellular pathological tau.

Table 1 Representative AD drug candidates targeting tau and neuroinflammation under clinical study

Mediators of tau post-translational modifications

Various kinases are known to induce tau hyperphosphorylation, such as GSK-3β, a major tau kinase. These kinases are hyperactivated during the progression of AD and other tauopathies [19], indicating that the development of their mediators may be an effective treatment for these diseases. Two GSK-3β inhibitors, lithium and tideglusib, have been tested [213]. Forlenza et al. reported that long-term lithium treatment at subtherapeutic doses attenuates cognitive and functional decline in amnestic MCI (ClinicalTrials.gov Identifier: NCT01055392) [214]. However, this dose of lithium treatment still has safety issues, such as side effects and tolerability (NCT01055392, NCT00703677) [215]. Another clinical trial evaluating the safety and efficacy of a lower dose of lithium in patients with mild to severe AD just started in June 2022 and is estimated to be completed in June 2024 (NCT05423522). Tideglusib, a thiadiazolidinone derivative, acts as a non-ATP competitive GSK-3β inhibitor. Tideglusib has been reported to reduce tau phosphorylation and has anti-inflammatory effects in animal models [216, 217]. The clinical studies in patients with mild-to-moderate AD (NCT01350362, NCT00948259) or PSP (NCT01049399) patients treated with tideglusib were negative on the clinical outcomes [218]; however, a subgroup analysis of PSP patients showed reduced brain atrophy [219].

There is a reciprocal relationship between O-GlcNAcylation and tau phosphorylation [43]. Inhibitors of the O-GlcNAcase enzyme (OGA)-like thiamet G increase tau glycosylation, reduce NFTs and neuronal cell death, improve motor behavior, and prolong survival of tau transgenic mice [174, 220, 221]. Three OGA inhibitors, ASN90, ASN51, and LY3372689, are currently in clinical trials. They have shown well safety and tolerance in phase 1 clinical trials [222, 223]. Phase 2/3 clinical trials of these inhibitors for the treatment of AD and PSP are ongoing (NCT05063539, [223]). Salsalate, which we discussed above, induces tau deacetylation. A clinical trial has shown that it does not affect disease progression in PSP patients (NCT02422485). A phase 1 clinical trial testing the safety, tolerability, and cognitive ability of salsalate in patients with mild to moderate AD (NCT03277573) has not yet published the results.

Anti-tau immunotherapy

In addition to the small molecule drugs targeting tau post-translational modifications discussed above, active and passive immunotherapies have been developed to neutralize and clear toxic tau species to reduce tau phosphorylation, aggregation, and dissemination. Active immunization, such as vaccination, can cause the body to produce an antibody-like response. AADvac1 and ACI-35/ACI-35.030 are anti-tau active vaccines for AD and other tauopathies currently in clinical trials [224, 225]. AADvac1 is a synthetic peptide corresponding to amino acids 294 to 305 of the tau sequence, and ACI-35 consists of 16 copies of a synthetic tau fragment phosphorylated at Ser396 and Ser404 sites [225]. Phase 1 clinical data show that AADvac1 has well safety, tolerability, and immunogenicity in patients with mild to moderate AD (NCT02031198, NCT01850238) [226]. However, the treatment of this vaccine did not slow cognitive and functional decline in mild AD patients in the phase 2 clinical trial (ADAMANT, NCT02579252), although it shows the high immunogenicity [227]. A phase 1 pilot trial investigating the effect of AADvac1 in patients with non-fluent primary progressive aphasia (AIDA) has yet to publish results (NCT03174886). ACI-35 treatment was reported to produce a weak immune response in a phase 1 study [228]. Therefore, a redesigned version, ACI-35.030, was indicated to elicit a stronger immune response [228]. A phase 1/2 clinical trial to test its safety and immunogenicity in patients with early AD is undergoing and expected to be completed in 2023 (NCT04445831).

Another immunotherapeutic approach is administering preformed anti-tau antibodies direct against different tau epitopes, also known as passive immunotherapy. Passive immune antibody drugs have the advantages of low risk of adverse effects to immunogenicity and more specificity for targeted epitopes. A total of 12 tau antibodies have entered clinical trials, but half of tau antibody trials have been terminated due to poor clinical efficacy for AD or PSP. These antibodies include gosuranemab [229], tilavonemab [230], zagotenemab [231], semorinemab [232], RG7345 [233], and BIIB076 [234]. These antibodies mainly bind the N- or C-terminal epitopes of tau. Recent clinical evidence indicates that antibodies targeting the microtubule-binding region (MTBR, residues 224 to 369) or phosphorylated epitopes around the center of tau are more likely to prevent the propagation of pathogenic aggregated tau [235, 236]. Antibodies binding MTBR such as E2814 [237] and antibodies targeting phosphorylated epitopes of MTBR or mid-domain of tau, including JNJ-63733657 [238], Lu AF87908 [239], PNT001 [225], bepranemab (UCB0107) [240], and APNmAb005 [241] have been shown to prevent tau pathology and dissemination in preclinical animal experiments. The antibody Lu AF87908 also regulates the uptake and lysosomal function to clear pathological tau by interacting with the IgG antibody receptor FcγR in primary microglia cultures [242], indicating that anti-tau antibodies may induce microglial clearance of tau and reduce tau propagation. Furthermore, the antibodies bepranemab, semorinemab, JNJ-63733657, E2814, and PNT001 showed good safety and tolerability in the phase 1 clinical trial (NCT03605082, NCT02820896, NCT03689153, NCT04231513, and NCT04096287). The phase 2/3 clinical trials to test the efficacy of E2814, bepranemab, semorinemab, and JNJ-63733657 in patients with AD or other tauopathies are ongoing (NCT04971733, NCT04867616, NCT03828747, and NCT04619420). Since E2814 is designed to target pathologic tau and can attenuate aggregated tau spreading in preclinical experiments [237], and lecanemab, an anti-Aβ antibody approved through the FDA accelerated approved process for AD treatment, can reduce the brain amyloid and slow the rate of cognitive decline in patients with early AD [10]. A phase 2/3 clinical study is underway to evaluate the cognition and clinical efficacy of E2814 alone, lecanemab alone, and the combination of these two drugs in participants with autosomal dominant AD (DIAN-TU, NCT05269394).

Therapeutically targeting tau through modulating neuroinflammation

A total of more than 50 inflammation-related agents have entered clinical trials for AD treatment, and 22 are currently undergoing mainly targeting the inflammatory (especially neuroinflammatory) response process that may result in neurodegeneration, such as regulating the function of microglia and the immune system, the activity of inflammatory response-related kinases/pathway, and the expression and release of pro-inflammatory factors. Here, we will mainly review neuroinflammation-related drugs in clinical trials that may also have modulatory effects on tau pathology (Table 1).

As mentioned above, p38α MAPK is a key kinase involved in microglia-induced neuroinflammation leading to tau pathology [135, 137], and this kinase can also directly regulate tau phosphorylation [39]. Recently, the p38α inhibitors such as neflamapimod have attracted attention as potential treatments for neurodegenerative diseases, including AD. A multi-center phase 2 clinical trial showed that although a 24-week treatment with neflamapimod did not ameliorate episodic memory in individuals with mild AD, neflamapimod treatment significantly reduces CSF total tau and tau phosphorylated at threonine 181 (pTau181) compared to the placebo group (NCT03402659). The results of this trial suggest that a longer study of neflamapimod at higher dose levels is needed to assess the effect on AD progression [243]. Notably, a phase 2a clinical study suggested that neflamapimod treatment improved cognitive and motor functions in patients with mild-to-moderate dementia with Lewy bodies (DLB) [244] (NCT04001517). These results indicate that inhibiting p38α MAPK to improve tau pathology may be one of the most promising strategies for AD treatment. Other p38α inhibitors, such as MW150 [245], MW151 [246], and MW189 (the intravenous formulation of MW151), all showed good safety and tolerability in phase 1 clinical trials (NCT04120233, NCT02942771) [247]. A phase 2 study of MW150 in patients with mild to moderate AD is underway (NCT05194163).

Nilotinib is a small molecule tyrosine kinase inhibitor with potent p38α inhibitory activity [248]. It modulates the immune profiles of glial cells and induces phosphorylated tau clearance, although not more efficiently than Aβ [249, 250]. A phase 2 clinical trial in 37 patients with mild to moderate AD (NCT02947893) showed that nilotinib was well-tolerated and reduced CSF pTau181 at 6 months compared to the placebo group [251].

NE3107, which reached phase 3 (NCT04669028), is a derivative of β-androstenediol. It binds to ERK and inhibits the activation of ERK/NF-κB signaling. NE3107 orally enters the brain. It has anti-neuroinflammatory and insulin-sensitizing properties, which make it attractive in AD treatment [252, 253]. A phase 2 clinical study presented at the 2022 clinical trials on Alzheimer’s disease (CTAD) conference showed that NE3107 was associated with improvement in CSF p-tau in patients with MCI or mild dementia [254]. A phase 3 trial to evaluate the safety and efficacy of this agent in subjects with mild to moderate probable AD (NCT04669028) started in August 2021 and is estimated to be completed in October 2023.

In addition, AZP2006, a small molecule to prevent the growth factor progranulin (PGRN) cleavage and promote its secretion, has also been reported to inhibit tau phosphorylation and neuroinflammation in a preclinical study [255]. The chronic treatment of AZP2006 attenuates the cognitive impairments and neuronal synaptic damage, accompanied by significant decreases in microglial activation, proinflammatory cytokine release, and tau hyperphosphorylation in the brains of AD and aging model mice. Further mechanistic studies demonstrated that AZP2006 binds to PSAP (a cofactor of PGRN) and inhibits the TLR9-driven signaling to reduce pro-inflammatory responses. These results indicate the potential of AZP2006 as a novel strategy for the treatment of AD and other tauopathies [255]. One phase 2 trial is ongoing to evaluate the safety, tolerability, pharmacokinetics, and effect of AZP2006 in patients with PSP (NCT04008355).

Another promising new therapy targeting tau pathology and neuroinflammation is the combination treatment of dasatinib and quercetin. Dasatinib is a cancer drug that inhibits Src tyrosine kinase. Quercetin inhibits the anti-apoptotic protein Bcl-xL and has anti-inflammatory activity. This drug combination eliminates the senescent cells (also known as “senolytic therapy”), reduces tau pathology, amyloid load, and neuroinflammation, and ameliorates cognitive impairment in preclinical studies [256, 257]. Dasatinib orally enters the brain, and the drug combination appears safe and tolerable in phase 1/2 clinical studies [258, 259]. Several clinical studies are underway to test this drug combination's efficacy in treating AD.

Sodium oligomannate (GV-971), a derivative of marine algae oligosaccharides, received conditional approval in China to treat mild to moderate AD in 2019 [260]. The preclinical data showed that GV-971 remodels the gut microbiota and the associated accumulation of phenylalanine and isoleucine, reduces microglial activation, immune responses, Aβ plaque deposition, and tau phosphorylation in the brain, and improves the cognitive impairment in 5xFAD mice [261]. GV-971 has also been demonstrated to reverse cognitive deficits in patients with mild to moderate AD in the phase 2/3 clinical trial in China (NCT02293915) [262]. Although the long-term efficacy and safety of GV-971 need to be further verified, these results highlight that multi-target drugs against multiple AD pathological changes may be a more potential therapeutic strategy.

Conclusions and perspectives

Over the past few decades, accumulated efforts in basic and clinical research in the AD field have improved our understanding of the multifactorial nature of AD pathogenesis. It is particularly encouraging that two DMT drugs targeting Aβ have recently received accelerated FDA approval for AD treatment. However, the efficacy of these drugs is controversial and needs further validation. The most likely reason for the failure of drugs targeting Aβ may be that Aβ pathology in AD does not always correlate with cognitive decline.

Tau has received increasing attention as a potential alternative therapeutic target, since evidence indicates that tau pathology is more associated with cognitive degradation. To date, there are no tau-focused drugs approved by FDA. Still, several agents targeting tau post-translational modification and dissemination have recently entered clinical trials for treating AD and other tauopathies. Accumulating evidence indicates that neuroinflammation may be the third pathological feature of AD and plays an integral role in AD pathogenesis and the promotion of cognitive impairment. In recent years, growing findings of fundamental research have demonstrated the complex interplay between neuroinflammation and tau pathology, as summarized above. These findings suggest that in the early stage of AD, a moderate inflammatory response may alleviate tau pathology, such as activated microglia promoting the clearance of tau seeds. While in the middle and late stages of AD, sustained and increased inflammatory responses in glial cells and neurons are pivotal cellular drivers and regulators of the exacerbation of tau pathology, further contributing to its worsening by promoting inflammatory responses. This vicious circle aggravates the pathological progression of AD. There are temporal and spatial dynamic regulatory processes between tau pathology and inflammation as the disease progresses. Further elucidation of these dynamic regulatory mechanisms will provide essential insights into AD pathogenesis and drug development.

Furthermore, current studies indicate that targeting only a single therapeutic target may not be able to reverse the pathological process of AD. As mentioned above, multi-target therapies (including multi-target single drugs such as the p38α inhibitors and single or multiple target multi-drug combination such as the combination of dasatinib and quercetin) targeting tau pathology and neuroinflammation, or simultaneously targeting Aβ pathology, which is currently undergoing clinical trials, may offer hope of reversing the course of AD and even curing it. These therapeutic strategies have shown good safety and tolerability, and clinical trials of their therapeutic effects are underway. In summary, both basic research and clinical trials suggest that targeting multiple pathologies and precise treatment strategies will be the trend of future drug development for AD and other tauopathies. They will be more likely to bring breakthroughs in the treatment of these diseases.

Availability of data and materials

All relevant data are included in this review.

Abbreviations

AD:

Alzheimer’s disease

Aβ:

β-Amyloid

ApoE4:

Apolipoprotein E4

cGAS:

Cyclic GMP–AMP synthase

CMKLR1:

Chemerin chemokine-like receptor 1

CX3CL1:

Chemokine (C–X3–C motif) ligand 1

CX3CR1:

CX3C motif chemokine receptor 1

FPR2:

Formyl peptide receptor 2

GSK-3β:

Glycogen synthase kinase-3 beta

IL-1β:

Interleukin-1β

HSPGs:

Heparan sulfate proteoglycans

LRP1:

Low-density lipoprotein receptor-related protein 1

MAPK:

Mitogen-activated protein kinase

NF-κB:

Nuclear factor kappa B

NFTs:

Neurofibrillary tangles

NLRP3:

NLR family pyrin domain-containing protein 3

P2RX7:

P2X purinoceptor 7

PHFs:

Paired helical filaments

STING:

Stimulator of interferon genes

TNF-α:

Tumor necrosis factor α

TREM2:

Triggering receptor expressed on myeloid cells 2

References

  1. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A, Cohen-Mansfield J, Cooper C, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396:413–46.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Dementia. https://www.who.int/news-room/fact-sheets/detail/dementia. Accessed 10 Apr 2023.

  3. Knopman DS, Amieva H, Petersen RC, Chetelat G, Holtzman DM, Hyman BT, Nixon RA, Jones DT. Alzheimer disease. Nat Rev Dis Primers. 2021;7:33.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Iqbal K, Liu F, Gong CX. Tau and neurodegenerative disease: the story so far. Nat Rev Neurol. 2016;12:13.

    Article  Google Scholar 

  5. Leng FD, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17:157–72.

    Article  PubMed  Google Scholar 

  6. Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol Neurodegener. 2020;15:40.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Cacace R, Sleegers K, Van Broeckhoven C. Molecular genetics of early-onset Alzheimer’s disease revisited. Alzheimers Dement. 2016;12:733–48.

    Article  PubMed  Google Scholar 

  8. Kantarci K. 2021 marks a new era for Alzheimer’s therapeutics. Lancet Neurol. 2022;21:3–4.

    Article  PubMed  Google Scholar 

  9. Mullard A. FDA approval for Biogen’s aducanumab sparks Alzheimer disease firestorm. Nat Rev Drug Discov. 2021;20:496–496.

    PubMed  Google Scholar 

  10. van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, Kanekiyo M, Li D, Reyderman L, Cohen S, et al. Lecanemab in early Alzheimer’s disease. N Engl J Med. 2023;388:9–21.

    Article  PubMed  Google Scholar 

  11. Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology. 1992;42:631–631.

    Article  CAS  PubMed  Google Scholar 

  12. GomezIsla T, Hollister R, West H, Mui S, Growdon JH, Petersen RC, Parisi JE, Hyman BT. Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer’s disease. Ann Neurol. 1997;41:17–24.

    Article  CAS  PubMed  Google Scholar 

  13. Gong P, Chen YQ, Lin AH, Zhang HB, Zhang Y, Ye RD, Yu Y. p47(phox) deficiency improves cognitive impairment and attenuates tau hyperphosphorylation in mouse models of AD. Alzheimers Res Ther. 2020;12:18.

    Article  Google Scholar 

  14. Zhang HB, Lin AH, Gong P, Chen YQ, Ye RD, Qian F, Zhang Y, Yu Y. The chemokine-like receptor 1 deficiency improves cognitive deficits of AD mice and attenuates tau hyperphosphorylation via regulating tau seeding. J Neurosci. 2020;40:6991–7007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bettcher BM, Tansey MG, Dorothee G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease—a research prospectus. Nat Rev Neurol. 2021;17:689–701.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Kidd M. Paired helical filaments in electron microscopy of Alzheimers disease. Nature. 1963;197:192–200.

    Article  CAS  PubMed  Google Scholar 

  17. Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. Protein factor essential for microtubule assembly. Proc Natl Acad Sci USA. 1975;72:1858–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Strang KH, Golde TE, Giasson BI. MAPT mutations, tauopathy, and mechanisms of neurodegeneration. Lab Invest. 2019;99:912–28.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol. 2017;133:665–704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Panda D, Samuel JC, Massie M, Feinstein SC, Wilson L. Differential regulation of microtubule dynamics by three- and four-repeat tau: Implications for the onset of neurodegenerative disease. Proc Natl Acad Sci USA. 2003;100:9548–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Goedert M. Tau gene mutations and their effects. Mov Disord. 2005;20:S45–52.

    Article  PubMed  Google Scholar 

  22. Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, Pickering-Brown S, Chakraverty S, Isaacs A, Grover A, et al. Association of missense and 5 ’-splice-site mutations in tau with the inherited dementia FTDP-17. Nature. 1998;393:702–5.

    Article  CAS  PubMed  Google Scholar 

  23. Hoglinger GU, Respondek G, Kovacs GG. New classification of tauopathies. Revue Neurologique. 2018;174:664–8.

    Article  CAS  PubMed  Google Scholar 

  24. Götz J, Halliday G, Nisbet RM. Molecular pathogenesis of the tauopathies. Annu Rev Pathol. 2019;14:239–61.

    Article  PubMed  Google Scholar 

  25. Hyman B. All the tau we cannot see. Annu Rev Med. 2023;74:503–14.

    Article  PubMed  Google Scholar 

  26. Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McGowan E, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Fox LM, William CM, Adamowicz DH, Pitstick R, Carlson GA, Spires-Jones TL, Hyman BT. Soluble tau species, not neurofibrillary aggregates, disrupt neural system integration in a tau transgenic model. J Neuropathol Exp Neurol. 2011;70:588–95.

    Article  CAS  PubMed  Google Scholar 

  28. Polydoro M, Dzhala VI, Pooler AM, Nicholls SB, McKinney AP, Sanchez L, Pitstick R, Carlson GA, Staley KJ, Spires-Jones TL, Hyman BT. Soluble pathological tau in the entorhinal cortex leads to presynaptic deficits in an early Alzheimer’s disease model. Acta Neuropathol. 2014;127:257–70.

    Article  CAS  PubMed  Google Scholar 

  29. Rudinskiy N, Hawkes JM, Betensky RA, Eguchi M, Yamaguchi S, Spires-Jones TL, Hyman BT. Orchestrated experience-driven Arc responses are disrupted in a mouse model of Alzheimer’s disease. Nat Neurosci. 2012;15:1422–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Busche MA, Wegmann S, Dujardin S, Commins C, Schiantarelli J, Klickstein N, Kamath TV, Carlson GA, Nelken I, Hyman BT. Tau impairs neural circuits, dominating amyloid-beta effects, in Alzheimer models in vivo. Nat Neurosci. 2019;22:57–64.

    Article  CAS  PubMed  Google Scholar 

  31. Wegmann S, DeVos SL, Zeitler B, Marlen K, Bennett RE, Perez-Rando M, MacKenzie D, Yu Q, Commins C, Bannon RN, et al. Persistent repression of tau in the brain using engineered zinc finger protein transcription factors. Sci Adv. 2021. https://doi.org/10.1126/sciadv.abe1611.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Wang YP, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci. 2016;17:5–21.

    Article  PubMed  Google Scholar 

  33. Merrick SE, Trojanowski JQ, Lee VMY. Selective destruction of stable microtubules and axons by inhibitors of protein serine/threonine phosphatases in cultured human neurons (NT2N cells). J Neurosci. 1997;17:5726–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li CZ, Gotz J. Tau-based therapies in neurodegeneration: opportunities and challenges. Nat Rev Drug Discovery. 2017;16:863–83.

    Article  CAS  PubMed  Google Scholar 

  35. Kopke E, Tung YC, Shaikh S, Alonso AD, Iqbal K, Grundkeiqbal I. Microtubule-associated protein-tau - abnormal phosphorylation of a non-paired helical filament pool in Alzheimer-disease. J Biol Chem. 1993;268:24374–84.

    Article  CAS  PubMed  Google Scholar 

  36. Alonso AD, Mederlyova A, Novak M, Grundke-Iqbal I, Iqbal K. Promotion of hyperphosphorylation by frontotemporal dementia tau mutations. J Biol Chem. 2004;279:34873–81.

    Article  PubMed  Google Scholar 

  37. Leroy K, Yilmaz Z, Brion JP. Increased level of active GSK-3 beta in Alzheimer’s disease and accumulation in argyrophilic grains and in neurones at different stages of neurofibrillary degeneration. Neuropathol Appl Neurobiol. 2007;33:43–55.

    Article  CAS  PubMed  Google Scholar 

  38. Patrick GN, Zukerberg L, Nikolic M, de la Monte S, Dikkes P, Tsai LH. Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration. Nature. 1999;402:615–22.

    Article  CAS  PubMed  Google Scholar 

  39. Stefanoska K, Gajwani M, Tan ARP, Ahel HI, Asih PR, Volkerling A, Poljak A, Ittner A. Alzheimer’s disease: ablating single master site abolishes tau hyperphosphorylation. Sci Adv. 2022. https://doi.org/10.1126/sciadv.abl8809.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Holmes BB, Furman JL, Mahan TE, Yamasaki TR, Mirbaha H, Eades WC, Belaygorod L, Cairns NJ, Holtzman DM, Diamond MI. Proteopathic tau seeding predicts tauopathy in vivo. Proc Natl Acad Sci USA. 2014;111:E4376–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mudher A, Colin M, Dujardin S, Medina M, Dewachter I, Naini SMA, Mandelkow EM, Mandelkow E, Buee L, Goedert M, Brion JP. What is the evidence that tau pathology spreads through prion-like propagation? Acta Neuropathol Commun. 2017;5:20.

    Article  Google Scholar 

  42. Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A, Fraser G, Stalder AK, Beibel M, Staufenbiel M, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009;11:909-U325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong CX. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer’s disease. Proc Natl Acad Sci USA. 2004;101:10804–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Brunden KR, Trojanowski JQ, Lee VMY. Advances in tau-focused drug discovery for Alzheimer’s disease and related tauopathies. Nat Rev Drug Discovery. 2009;8:783–93.

    Article  CAS  PubMed  Google Scholar 

  45. Wang JZ, Grundke-Iqbal I, Iqbal K. Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer’s disease. Nat Med. 1996;2:871–5.

    Article  CAS  PubMed  Google Scholar 

  46. Min SW, Cho SH, Zhou YG, Schroeder S, Haroutunian V, Seeley WW, Huang EJ, Shen Y, Masliah E, Mukherjee C, et al. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron. 2010;67:953–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cohen TJ, Guo JL, Hurtado DE, Kwong LK, Mills IP, Trojanowski JQ, Lee VMY. The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun. 2011;2:9.

    Article  Google Scholar 

  48. Min SW, Chen X, Tracy TE, Li YQ, Zhou YG, Wang C, Shirakawa K, Minami SS, Defensor E, Mok SA, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med. 2015;21:1154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Irwin DJ, Cohen TJ, Grossman M, Arnold SE, McCarty-Wood E, Van Deerlin VM, Lee VMY, Trojanowski JQ. Acetylated tau neuropathology in sporadic and hereditary tauopathies. Am J Pathol. 2013;183:344–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Caballero B, Bourdenx M, Luengo E, Diaz A, Sohn PD, Chen X, Wang C, Juste YR, Wegmann S, Patel B, et al. Acetylated tau inhibits chaperone-mediated autophagy and promotes tau pathology propagation in mice. Nat Commun. 2021;12:2238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shin M-K, Vazquez-Rosa E, Koh Y, Dhar M, Chaubey K, Cintron-Perez CJ, Barker S, Miller E, Franke K, Noterman MF, et al. Reducing acetylated tau is neuroprotective in brain injury. Cell. 2021;184:2715–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lee S, Shea TB. Caspase-mediated truncation of tau potentiates aggregation. Int J Alzheimer’s Dis. 2012;2012: 731063.

    Google Scholar 

  53. Kondo A, Shahpasand K, Mannix R, Qiu JH, Moncaster J, Chen CH, Yao YD, Lin YM, Driver JA, Sun Y, et al. Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature. 2015;523:431-U118.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Funk KE, Thomas SN, Schafer KN, Cooper GL, Liao ZP, Clark DJ, Yang AJ, Kuret J. Lysine methylation is an endogenous post-translational modification of tau protein in human brain and a modulator of aggregation propensity. Biochemical Journal. 2014;462:77–88.

    Article  CAS  PubMed  Google Scholar 

  55. Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11:155–9.

    Article  CAS  PubMed  Google Scholar 

  56. Petkova AT, Leapman RD, Guo Z, Yau WM, Mattson MP, Tycko R. Self-propagating, molecular-level polymorphism in Alzheimer’s beta-amyloid fibrils. Science. 2005;307:262–5.

    Article  CAS  PubMed  Google Scholar 

  57. Braak H, Braak E. Diagnostic criteria for neuropathologic assessment of Alzheimer’s disease. Neurobiol Aging. 1997;18:S85–8.

    Article  CAS  PubMed  Google Scholar 

  58. Peeraer E, Bottelbergs A, Van Kolen K, Stancu IC, Vasconcelos B, Mahieu M, Duytschaever H, Donck LV, Torremans A, Sluydts E, et al. Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice. Neurobiol Dis. 2015;73:83–95.

    Article  CAS  PubMed  Google Scholar 

  59. Alonso AD, Beharry C, Corbo CP, Cohen LS. Molecular mechanism of prion-like tau-induced neurodegeneration. Alzheimers Dement. 2016;12:1090–7.

    Article  PubMed  Google Scholar 

  60. Takeda S, Wegmann S, Cho HS, Devos SL, Commins C, Roe AD, Nicholls SB, Carlson GA, Pitstick R, Nobuhara CK, et al. Neuronal uptake and propagation of a rare phosphorylated high-molecular-weight tau derived from Alzheimer’s disease brain. Nat Commun. 2015;6:15.

    Article  Google Scholar 

  61. Dai CL, Hu W, Tung YC, Liu F, Gong CX, Iqbal K. Tau passive immunization blocks seeding and spread of Alzheimer hyperphosphorylated tau-induced pathology in 3 x Tg-AD mice. Alzheimers Res Ther. 2018;10:14.

    Article  Google Scholar 

  62. Iba M, McBride JD, Guo JL, Zhang B, Trojanowski JQ, Lee VMY. 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. 2015;130:349–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Wegmann S, Maury EA, Kirk MJ, Saqran L, Roe A, DeVos SL, Nicholls S, Fan Z, Takeda S, Cagsal-Getkin O, et al. Removing endogenous tau does not prevent tau propagation yet reduces its neurotoxicity. Embo j. 2015;34:3028–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Calafate S, Buist A, Miskiewicz K, Vijayan V, Daneels G, de Strooper B, de Wit J, Verstreken P, Moechars D. Synaptic contacts enhance cell-to-cell tau pathology propagation. Cell Rep. 2015;11:1176–83.

    Article  CAS  PubMed  Google Scholar 

  65. Polanco JC, Gotz J. Exosomal and vesicle-free tau seeds-propagation and convergence in endolysosomal permeabilization. FEBS J. 2022;289:6891–907.

    Article  CAS  PubMed  Google Scholar 

  66. Sokolow S, Henkins KM, Bilousova T, Gonzalez B, Vinters HV, Miller CA, Cornwell L, Poon WW, Gylys KH. Pre-synaptic C-terminal truncated tau is released from cortical synapses in Alzheimer’s disease. J Neurochem. 2015;133:368–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Perez M, Avila J, Hernandez F. Propagation of Tau via Extracellular Vesicles. Front Neurosci. 2019;13:7.

    Article  Google Scholar 

  68. McInnes J, Wierda K, Snellinx A, Bounti L, Wang YC, Stancu IC, Apostolo N, Gevaert K, Dewachter I, Spires-Jones TL, et al. Synaptogyrin-3 mediates presynaptic dysfunction induced by tau. Neuron. 2018;97:823.

    Article  CAS  PubMed  Google Scholar 

  69. Calafate S, Flavin W, Verstreken P, Moechars D. Loss of bin1 promotes the propagation of tau pathology. Cell Rep. 2016;17:931–40.

    Article  CAS  PubMed  Google Scholar 

  70. Leyns CEG, Gratuze M, Narasimhan S, Jain N, Koscal LJ, Jiang H, Manis M, Colonna M, Lee VMY, Ulrich JD, Holtzman DM. TREM2 function impedes tau seeding in neuritic plaques. Nat Neurosci. 2019;22:1217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Christianson HC, Belting M. Heparan sulfate proteoglycan as a cell-surface endocytosis receptor. Matrix Biol. 2014;35:51–5.

    Article  CAS  PubMed  Google Scholar 

  72. Meldolesi J. Extracellular vesicles (exosomes and ectosomes) play key roles in the pathology of brain diseases. Mol Biomed. 2021;2:18.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Saman S, Kim W, Raya M, Visnick Y, Miro S, Jackson B, McKee AC, Alvarez VE, Lee NCY, Hall GF. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J Biol Chem. 2012;287:3842–9.

    Article  CAS  PubMed  Google Scholar 

  74. Rauch JN, Luna G, Guzman E, Audouard M, Challis C, Sibih YE, Leshuk C, Hernandez I, Wegmann S, Hyman BT, et al. LRP1 is a master regulator of tau uptake and spread. Nature. 2020;580:381.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang Y, Balaji V, Kaniyappan S, Kruger L, Irsen S, Tepper K, Chandupatla R, Maetzler W, Schneider A, Mandelkow E, Mandelkow EM. The release and trans-synaptic transmission of Tau via exosomes. Mol Neurodegener. 2017;12:25.

    Article  Google Scholar 

  76. Rajendran L, Honsho M, Zahn TR, Keller P, Geiger KD, Verkade P, Simons K. Alzheimer’s disease beta-amyloid peptides are released in association with exosomes. Proc Natl Acad Sci USA. 2006;103:11172–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Fiandaca MS, Kapogiannis D, Mapstone M, Boxer A, Eitan E, Schwartz JB, Abner EL, Petersen RC, Federoff HJ, Miller BL, Goetzl EJ. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: a case-control study. Alzheimers & Dementia. 2015;11:600–7.

    Article  Google Scholar 

  78. Wu JW, Hussaini SA, Bastille IM, Rodriguez GA, Mrejeru A, Rilett K, Sanders DW, Cook C, Fu H, Boonen RA, et al. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat Neurosci. 2016;19:1085–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Agosta F, Dalla Libera D, Spinelli EG, Finardi A, Canu E, Bergami A, Chiavetto LB, Baronio M, Comi G, Martino G, et al. Myeloid microvesicles in cerebrospinal fluid are associated with myelin damage and neuronal loss in mild cognitive impairment and alzheimer disease. Ann Neurol. 2014;76:813–25.

    Article  CAS  PubMed  Google Scholar 

  80. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, Wolozin B, Butovsky O, Kugler S, Ikezu T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18:1584–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Sanders DW, Kaufman SK, DeVos SL, Sharma AM, Mirbaha H, Li AM, Barker SJ, Foley AC, Thorpe JR, Serpell LC, et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron. 2014;82:1271–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Santello M, Toni N, Volterra A. Astrocyte function from information processing to cognition and cognitive impairment. Nat Neurosci. 2019;22:154–66.

    Article  CAS  PubMed  Google Scholar 

  83. Pearce MMP, Spartz EJ, Hong W, Luo L, Kopito RR. Prion-like transmission of neuronal huntingtin aggregates to phagocytic glia in the Drosophila brain. Nat Commun. 2015;6:6768.

    Article  CAS  PubMed  Google Scholar 

  84. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (NY). 2018;4:575–90.

    Article  Google Scholar 

  85. Meraz-Ríos MA, Toral-Rios D, Franco-Bocanegra D, Villeda-Hernández J, Campos-Peña V. Inflammatory process in Alzheimer’s disease. Front Integr Neurosci. 2013;7:59.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308:1314–8.

    Article  CAS  PubMed  Google Scholar 

  87. Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017;23:1018–27.

    Article  CAS  PubMed  Google Scholar 

  88. Yu Y, Ye RD. Microglial Abeta receptors in Alzheimer’s disease. Cell Mol Neurobiol. 2015;35:71–83.

    Article  CAS  PubMed  Google Scholar 

  89. Sarlus H, Heneka MT. Microglia in Alzheimer’s disease. J Clin Investig. 2017;127:3240–9.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Feng WX, Zhang YL, Wang Z, Xu HR, Wu T, Marshall C, Gao JY, Xiao M. Microglia prevent beta-amyloid plaque formation in the early stage of an Alzheimer’s disease mouse model with suppression of glymphatic clearance. Alzheimers Res Ther. 2020;12:125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. J Neurosci. 2008;28:8354–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Michelucci A, Heurtaux T, Grandbarbe L, Morga E, Heuschling P. Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: effects of oligomeric and fibrillar amyloid-beta. J Neuroimmunol. 2009;210:3–12.

    Article  CAS  PubMed  Google Scholar 

  93. Avila-Munoz E, Arias C. When astrocytes become harmful: functional and inflammatory responses that contribute to Alzheimer’s disease. Ageing Res Rev. 2014;18:29–40.

    Article  CAS  PubMed  Google Scholar 

  94. Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119:7–35.

    Article  PubMed  Google Scholar 

  95. Carrero I, Gonzalo MR, Martin B, Sanz-Anquela JM, Arevalo-Serrano J, Gonzalo-Ruiz A. Oligomers of beta-amyloid protein (A beta 1–42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain. Exp Neurol. 2012;236:215–27.

    Article  CAS  PubMed  Google Scholar 

  96. Garwood CJ, Pooler AM, Atherton J, Hanger DP, Noble W. Astrocytes are important mediators of Abeta-induced neurotoxicity and tau phosphorylation in primary culture. Cell Death Dis. 2011;2: e167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Allaman I, Belanger M, Magistretti PJ. Astrocyte-neuron metabolic relationships: for better and for worse. Trends Neurosci. 2011;34:76–87.

    Article  CAS  PubMed  Google Scholar 

  98. Blum-Degen D, Mueller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin-1-beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci Lett. 1995;202:17–20.

    Article  CAS  PubMed  Google Scholar 

  99. Tarkowski E, Issa R, Sjogren M, Wallin A, Blennow K, Tarkowski A, Kumar P. Increased intrathecal levels of the angiogenic factors VEGF and TGF-beta in Alzheimer’s disease and vascular dementia. Neurobiol Aging. 2002;23:237–43.

    Article  CAS  PubMed  Google Scholar 

  100. Jiang H, Hampel H, Prvulovic D, Wallin A, Blennow K, Li RN, Shen Y. Elevated CSF levels of TACE activity and soluble TNF receptors in subjects with mild cognitive impairment and patients with Alzheimer’s disease. Mol Neurodegener. 2011;6:8.

    Article  Google Scholar 

  101. Schuitemaker A, Dik MG, Veerhuis R, Scheltens P, Schoonenboom NSM, Hack CE, Blankenstein MA, Jonker C. Inflammatory markers in AD and MCI patients with different biomarker profiles. Neurobiol Aging. 2009;30:1885–9.

    Article  CAS  PubMed  Google Scholar 

  102. McGeer PL, Rogers J, McGeer EG. Inflammation, antiinflammatory agents, and Alzheimer’s disease: the last 22 years. J Alzheimers Dis. 2016;54:853–7.

    Article  PubMed  Google Scholar 

  103. Nordengen K, Kirsebom BE, Henjum K, Selnes P, Gisladottir B, Wettergreen M, Torsetnes SB, Grontvedt GR, Waterloo KK, Aarsland D, et al. Glial activation and inflammation along the Alzheimer’s disease continuum. J Neuroinflammation. 2019;16:13.

    Article  Google Scholar 

  104. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Blasko I, Grubeck-Loebenstein B. Role of the immune system in the pathogenesis, prevention and treatment of Alzheimer’s disease. Drugs Aging. 2003;20:101–13.

    Article  CAS  PubMed  Google Scholar 

  106. Morris JK, Honea RA, Vidoni ED, Swerdlow RH, Burns JM. Is Alzheimer’s disease a systemic disease? BBA-Mol Basis Dis. 2014;1842:1340–9.

    Article  CAS  Google Scholar 

  107. Walker KA, Ficek BN, Westbrook R. Understanding the role of systemic inflammation in Alzheimer’s disease. ACS Chem Neurosci. 2019;10:3340–2.

    Article  CAS  PubMed  Google Scholar 

  108. Vlad SC, Miller DR, Kowall NW, Felson DT. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology. 2008;70:1672–7.

    Article  CAS  PubMed  Google Scholar 

  109. Gasparini L, Ongini E, Wenk G. Non-steroidal anti-inflammatory drugs (NSAIDs) in Alzheimer’s disease: old and new mechanisms of action. J Neurochem. 2004;91:521–36.

    Article  CAS  PubMed  Google Scholar 

  110. Etminan M, Gill S, Samii A. Effect of non-steroidal anti-inflammatory drugs on risk of Alzheimer’s disease: systematic review and meta-analysis of observational studies. BMJ. 2003;327:128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Pasqualetti P, Bonomini C, Dal Forno G, Paulon L, Sinforiani E, Marra C, Zanetti O, Rossini PM. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer’s disease. Aging Clin Exp Res. 2009;21:102–10.

    Article  CAS  PubMed  Google Scholar 

  112. Thal LJ, Ferris SH, Kirby L, Block GA, Lines CR, Yuen E, Assaid C, Nessly ML, Norman BA, Baranak CC, et al. A randomized, double-blind, study of rofecoxib in patients with mild cognitive impairment. Neuropsychopharmacology. 2005;30:1204–15.

    Article  CAS  PubMed  Google Scholar 

  113. Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, Cribbs DH, LaFerla FM. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal β-catenin pathway function in an Alzheimer’s disease model. J Immunol. 2011;187:6539–49.

    Article  CAS  PubMed  Google Scholar 

  114. Leyns CEG, Holtzman DM. Glial contributions to neurodegeneration in tauopathies. Mol Neurodegener. 2017;12:50.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Laurent C, Buee L, Blum D. Tau and neuroinflammation: What impact for Alzheimer’s disease and tauopathies? Biomed J. 2018;41:21–33.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Felsky D, Roostaei T, Nho K, Risacher SL, Bradshaw EM, Petyuk V, Schneider JA, Saykin A, Bennett DA, De Jager PL. Neuropathological correlates and genetic architecture of microglial activation in elderly human brain. Nat Commun. 2019;10:409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liu J, Wang D, Li SQ, Yu Y, Ye RD. Suppression of LPS-induced tau hyperphosphorylation by serum amyloid A. J Neuroinflammation. 2016;13:15.

    Article  Google Scholar 

  118. Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J Neurosci. 2005;25:8843–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, Lah JJ, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med. 2013;368:107–16.

    Article  CAS  PubMed  Google Scholar 

  120. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368:117–27.

    Article  CAS  PubMed  Google Scholar 

  121. Zhao Y, Wu X, Li X, Jiang LL, Gui X, Liu Y, Sun Y, Zhu B, Pina-Crespo JC, Zhang M, et al. TREM2 is a receptor for beta-amyloid that mediates microglial function. Neuron. 2018;97(1023–1031): e1027.

    Google Scholar 

  122. Takahashi K, Rochford CD, Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J Exp Med. 2005;201:647–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Sessa G, Podini P, Mariani M, Meroni A, Spreafico R, Sinigaglia F, Colonna M, Panina P, Meldolesi J. Distribution and signaling of TREM2/DAP12, the receptor system mutated in human polycystic lipomembraneous osteodysplasia with sclerosing leukoencephalopathy dementia. Eur J Neurosci. 2004;20:2617–28.

    Article  PubMed  Google Scholar 

  124. Lill CM, Rengmark A, Pihlstrom L, Fogh I, Shatunov A, Sleiman PM, Wang LS, Liu T, Lassen CF, Meissner E, et al. The role of TREM2 R47H as a risk factor for Alzheimer’s disease, frontotemporal lobar degeneration, amyotrophic lateral sclerosis, and Parkinson’s disease. Alzheimers Dement. 2015;11:1407–16.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Piccio L, Deming Y, Del-Aguila JL, Ghezzi L, Holtzman DM, Fagan AM, Fenoglio C, Galimberti D, Borroni B, Cruchaga C. Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathol. 2016;131:925–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Bemiller SM, McCray TJ, Allan K, Formica SV, Xu G, Wilson G, Kokiko-Cochran ON, Crish SD, Lasagna-Reeves CA, Ransohoff RM, et al. TREM2 deficiency exacerbates tau pathology through dysregulated kinase signaling in a mouse model of tauopathy. Mol Neurodegener. 2017;12:74.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Leyns CEG, Ulrich JD, Finn MB, Stewart FR, Koscal LJ, Remolina Serrano J, Robinson GO, Anderson E, Colonna M, Holtzman DM. TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci U S A. 2017;114:11524–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Lee SH, Meilandt WJ, Xie L, Gandham VD, Ngu H, Barck KH, Rezzonico MG, Imperio J, Lalehzadeh G, Huntley MA, et al. Trem2 restrains the enhancement of tau accumulation and neurodegeneration by beta-amyloid pathology. Neuron. 2021;109(1283–1301): e1286.

    Google Scholar 

  129. Venkatakrishnan AJ, Deupi X, Lebon G, Tate CG, Schertler GF, Babu MM. Molecular signatures of G-protein-coupled receptors. Nature. 2013;494:185–94.

    Article  CAS  PubMed  Google Scholar 

  130. Liu Z, Condello C, Schain A, Harb R, Grutzendler J. CX3CR1 in microglia regulates brain amyloid deposition through selective protofibrillar amyloid-beta phagocytosis. J Neurosci. 2010;30:17091–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, Ransohoff RM, Lamb BT. CX3CR1 deficiency alters microglial activation and reduces beta-amyloid deposition in two Alzheimer’s disease mouse models. Am J Pathol. 2010;177:2549–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Puntambekar SS, Moutinho M, Lin PBC, Jadhav V, Tumbleson-Brink D, Balaji A, Benito MA, Xu GX, Oblak A, Lasagna-Reeves CA, et al. CX3CR1 deficiency aggravates amyloid driven neuronal pathology and cognitive decline in Alzheimer’s disease. Mol Neurodegener. 2022;17:21.

    Article  Google Scholar 

  133. Lee S, Xu G, Jay TR, Bhatta S, Kim KW, Jung S, Landreth GE, Ransohoff RM, Lamb BT. Opposing effects of membrane-anchored CX3CL1 on amyloid and tau pathologies via the p38 MAPK pathway. J Neurosci. 2014;34:12538–46.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Fuhrmann M, Bittner T, Jung CK, Burgold S, Page RM, Mitteregger G, Haass C, LaFerla FM, Kretzschmar H, Herms J. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat Neurosci. 2010;13:411–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, Lamb BT, Bhaskar K. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738–55.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Fan Q, He W, Gayen M, Benoit MR, Luo X, Hu X, Yan R. Activated CX3CL1/Smad2 signals prevent neuronal loss and alzheimer’s tau pathology-mediated cognitive dysfunction. J Neurosci. 2020;40:1133–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of Tau Pathology by the Microglial Fractalkine Receptor. Neuron. 2010;68:19–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Uddin MS, Kabir MT, Al Mamun A, Abdel-Daim MM, Barreto GE, Ashraf GM. APOE and Alzheimer’s disease: evidence mounts that targeting APOE4 may combat Alzheimer’s pathogenesis. Mol Neurobiol. 2019;56:2450–65.

    Article  CAS  PubMed  Google Scholar 

  139. Brecht WJ, Harris FM, Chang S, Tesseur I, Yu GQ, Xu Q, Dee Fish J, Wyss-Coray T, Buttini M, Mucke L, et al. Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice. J Neurosci. 2004;24:2527–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Jablonski AM, Warren L, Usenovic M, Zhou H, Sugam J, Parmentier-Batteur S, Voleti B. Astrocytic expression of the Alzheimer’s disease risk allele, ApoEepsilon4, potentiates neuronal tau pathology in multiple preclinical models. Sci Rep. 2021;11:3438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Saroja SR, Gorbachev K, Julia TCW, Goate AM, Pereira AC. Astrocyte-secreted glypican-4 drives APOE4-dependent tau hyperphosphorylation. Proc Natl Acad Sci U S A. 2022. https://doi.org/10.1073/pnas.2108870119.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Wang C, Xiong M, Gratuze M, Bao X, Shi Y, Andhey PS, Manis M, Schroeder C, Yin Z, Madore C, et al. Selective removal of astrocytic APOE4 strongly protects against tau-mediated neurodegeneration and decreases synaptic phagocytosis by microglia. Neuron. 2021;109(1657–1674): e1657.

    Article  Google Scholar 

  143. Shi Y, Yamada K, Liddelow SA, Smith ST, Zhao L, Luo W, Tsai RM, Spina S, Grinberg LT, Rojas JC, et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature. 2017;549:523–7.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Kovacs GG, Ferrer I, Grinberg LT, Alafuzoff I, Attems J, Budka H, Cairns NJ, Crary JF, Duyckaerts C, Ghetti B, et al. Aging-related tau astrogliopathy (ARTAG): harmonized evaluation strategy. Acta Neuropathol. 2016;131:87–102.

    Article  CAS  PubMed  Google Scholar 

  145. Richetin K, Steullet P, Pachoud M, Perbet R, Parietti E, Maheswaran M, Eddarkaoui S, Begard S, Pythoud C, Rey M, et al. Tau accumulation in astrocytes of the dentate gyrus induces neuronal dysfunction and memory deficits in Alzheimer’s disease. Nat Neurosci. 2020;23:1567–79.

    Article  CAS  PubMed  Google Scholar 

  146. Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM. International union of basic and clinical pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev. 2009;61:119–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Kennedy AJ, Davenport AP. International union of basic and clinical pharmacology CIII: chemerin receptors CMKLR1 (Chemerin(1)) and GPR1 (Chemerin(2)) nomenclature, pharmacology, and function. Pharmacol Rev. 2018;70:174–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Le Y, Gong W, Tiffany HL, Tumanov A, Nedospasov S, Shen W, Dunlop NM, Gao JL, Murphy PM, Oppenheim JJ, Wang JM. Amyloid (beta)42 activates a G-protein-coupled chemoattractant receptor, FPR-like-1. J Neurosci. 2001;21:RC123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Peng L, Yu Y, Liu J, Li S, He H, Cheng N, Ye RD. The chemerin receptor CMKLR1 is a functional receptor for amyloid-beta peptide. J Alzheimers Dis. 2015;43:227–42.

    Article  CAS  PubMed  Google Scholar 

  150. Zhu Y, Lin X, Zong X, Han S, Wang M, Su Y, Ma L, Chu X, Yi C, Zhao Q, Wu B. Structural basis of FPR2 in recognition of Abeta(42) and neuroprotection by humanin. Nat Commun. 2022;13:1775.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Medeiros R, Kitazawa M, Passos GF, Baglietto-Vargas D, Cheng D, Cribbs DH, LaFerla FM. Aspirin-triggered lipoxin A4 stimulates alternative activation of microglia and reduces Alzheimer disease-like pathology in mice. Am J Pathol. 2013;182:1780–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Zhang H, Wang D, Gong P, Lin A, Zhang Y, Ye RD, Yu Y. Formyl peptide receptor 2 deficiency improves cognition and attenuates tau hyperphosphorylation and astrogliosis in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2019;67:169–79.

    Article  CAS  PubMed  Google Scholar 

  153. Schroder N, Schaffrath A, Welter JA, Putzka T, Griep A, Ziegler P, Brandt E, Samer S, Heneka MT, Kaddatz H, et al. Inhibition of formyl peptide receptors improves the outcome in a mouse model of Alzheimer disease. J Neuroinflammation. 2020;17:131.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Trojan E, Tylek K, Schroder N, Kahl I, Brandenburg LO, Mastromarino M, Leopoldo M, Basta-Kaim A, Lacivita E. The N-formyl peptide receptor 2 (FPR2) agonist MR-39 improves ex vivo and in vivo amyloid beta (1–42)-induced neuroinflammation in mouse models of Alzheimer’s disease. Mol Neurobiol. 2021;58:6203–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Chen Y, Liu Z, Gong P, Zhang H, Chen Y, Yao S, Li W, Zhang Y, Yu Y. The chemerin/CMKLR1 axis is involved in the recruitment of microglia to abeta deposition through p38 MAPK pathway. Int J Mol Sci. 2022;23:9041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Hamlett ED, Hjorth E, Ledreux A, Gilmore A, Schultzberg M, Granholm AC. RvE1 treatment prevents memory loss and neuroinflammation in the Ts65Dn mouse model of Down syndrome. Glia. 2020;68:1347–60.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Cash JL, Hart R, Russ A, Dixon JP, Colledge WH, Doran J, Hendrick AG, Carlton MB, Greaves DR. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J Exp Med. 2008;205:767–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Lei Z, Lu Y, Bai X, Jiang Z, Yu Q. Chemerin-9 peptide enhances memory and ameliorates abeta(1–42)-induced object memory impairment in mice. Biol Pharm Bull. 2020;43:272–83.

    Article  CAS  PubMed  Google Scholar 

  159. Ising C, Venegas C, Zhang SS, Scheiblich H, Schmidt SV, Vieira-Saecker A, Schwartz S, Albasset S, McManus RM, Tejera D, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Heneka MT, McManus RM, Latz E. Inflammasome signalling in brain function and neurodegenerative disease. Nat Rev Neurosci. 2018;19:610–21.

    Article  CAS  PubMed  Google Scholar 

  161. Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23:1605–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Ghosh S, Wu MD, Shaftel SS, Kyrkanides S, LaFerla FM, Olschowka JA, O’Banion MK. Sustained interleukin-1beta overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J Neurosci. 2013;33:5053–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Sen T, Saha P, Jiang T, Sen N. Sulfhydration of AKT triggers Tau-phosphorylation by activating glycogen synthase kinase 3beta in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2020;117:4418–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Maphis N, Jiang SY, Xu GX, Kokiko-Cochran ON, Roy SM, Van Eldik LJ, Watterson DM, Lamb BT, Bhaskar K. Selective suppression of the alpha isoform of p38 MAPK rescues late-stage tau pathology. Alzheimers Res Ther. 2016;8:54.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Weston LL, Jiang S, Chisholm D, Jantzie LL, Bhaskar K. Interleukin-10 deficiency exacerbates inflammation-induced tau pathology. J Neuroinflammation. 2021;18:161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Li SQ, Yu Y, Han JZ, Wang D, Liu J, Qian F, Fan GH, Bucala R, Ye RD. Deficiency of macrophage migration inhibitory factor attenuates tau hyperphosphorylation in mouse models of Alzheimer’s disease. J Neuroinflammation. 2015;12:11.

    Article  CAS  Google Scholar 

  167. Zheng C, Zhou XW, Wang JZ. The dual roles of cytokines in Alzheimer’s disease: update on interleukins, TNF-alpha, TGF-beta and IFN-gamma. Transl Neurodegener. 2016;5:7.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Quintanilla RA, Orellana DI, Gonzalez-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res. 2004;295:245–57.

    Article  CAS  PubMed  Google Scholar 

  169. Zambrano A, Otth C, Maccioni RB, Concha II. IL-3 controls tau modifications and protects cortical neurons from neurodegeneration. Curr Alzheimer Res. 2010;7:615–24.

    Article  CAS  PubMed  Google Scholar 

  170. McAlpine CS, Park J, Griciuc A, Kim E, Choi SH, Iwamoto Y, Kiss MG, Christie KA, Vinegoni C, Poller WC, et al. Astrocytic interleukin-3 programs microglia and limits Alzheimer’s disease. Nature. 2021;595:701–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Losev Y, Frenkel-Pinter M, Abu-Hussien M, Viswanathan GK, Elyashiv-Revivo D, Geries R, Khalaila I, Gazit E, Segal D. Differential effects of putative N-glycosylation sites in human Tau on Alzheimer’s disease-related neurodegeneration. Cell Mol Life Sci. 2021;78:2231–45.

    Article  CAS  PubMed  Google Scholar 

  172. Zhu YP, Shan XY, Yuzwa SA, Vocadlo DJ. The emerging link between O-GlcNAc and Alzheimer disease. J Biol Chem. 2014;289:34472–81.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Losev Y, Paul A, Frenkel-Pinter M, Abu-Hussein M, Khalaila I, Gazit E, Segal D. Novel model of secreted human tau protein reveals the impact of the abnormal N-glycosylation of tau on its aggregation propensity. Sci Rep. 2019;9:10.

    Article  Google Scholar 

  174. Yuzwa SA, Shan X, Macauley MS, Clark T, Skorobogatko Y, Vosseller K, Vocadlo DJ. Increasing O-GlcNAc slows neurodegeneration and stabilizes tau against aggregation. Nat Chem Biol. 2012;8:393–9.

    Article  CAS  PubMed  Google Scholar 

  175. Pinho TS, Correia SC, Perry G, Ambrosio AF, Moreira PI. Diminished O-GlcNAcylation in Alzheimer’s disease is strongly correlated with mitochondrial anomalies. Biochim Biophys Acta Mol Basis Dis. 2019;1865:2048–59.

    Article  CAS  PubMed  Google Scholar 

  176. Park JS, Ji IJ, Kim DH, An HJ, Yoon SY. The Alzheimer’s disease-associated R47H variant of TREM2 has an altered glycosylation pattern and protein stability. Front Neurosci. 2016;10:618.

    PubMed  Google Scholar 

  177. Haukedal H, Freude KK. Implications of glycosylation in Alzheimer’s disease. Front Neurosci. 2021;14:18.

    Article  Google Scholar 

  178. Rebelo AL, Chevalier MT, Russo L, Pandit A. Role and therapeutic implications of protein glycosylation in neuroinflammation. Trends Mol Med. 2022;28:270–89.

    Article  CAS  PubMed  Google Scholar 

  179. Aubry S, Shin W, Crary JF, Lefort R, Qureshi YH, Lefebvre C, Califano A, Shelanski ML. Assembly and interrogation of Alzheimer’s disease genetic networks reveal novel regulators of progression. PLoS ONE. 2015;10: e0120352.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang XF, Yao TP. HDAC6 is a microtubule-associated deacetylase. Nature. 2002;417:455–8.

    Article  CAS  PubMed  Google Scholar 

  181. Trzeciakiewicz H, Ajit D, Tseng JH, Chen Y, Ajit A, Tabassum Z, Lobrovich R, Peterson C, Riddick NV, Itano MS, et al. An HDAC6-dependent surveillance mechanism suppresses tau-mediated neurodegeneration and cognitive decline. Nat Commun. 2020;11:5522.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Tseng JH, Xie L, Song S, Xie YM, Allen L, Ajit D, Hong JS, Chen X, Meeker RB, Cohen TJ. The deacetylase HDAC6 mediates endogenous neuritic tau pathology. Cell Rep. 2017;20:2169–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Shuttleworth SJ, Bailey SG, Townsend PA. Histone Deacetylase inhibitors: new promise in the treatment of immune and inflammatory diseases. Curr Drug Targets. 2010;11:1430–8.

    Article  CAS  PubMed  Google Scholar 

  184. Youn GS, Cho H, Kim D, Choi SY, Park J. Crosstalk between HDAC6 and Nox2-based NADPH oxidase mediates HIV-1 Tat-induced pro-inflammatory responses in astrocytes. Redox Biol. 2017;12:978–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Amro Z, Yool AJ, Collins-Praino LE. The potential role of glial cells in driving the prion-like transcellular propagation of tau in tauopathies. Brain Behav Immun. 2021;14: 100242.

    CAS  Google Scholar 

  186. Stancu I-C, Cremers N, Vanrusselt H, Couturier J, Vanoosthuyse A, Kessels S, Lodder C, Brone B, Huaux F, Octave J-N, et al. Aggregated Tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol. 2019;137:599–617.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, Savard M, Lussier FZ, Tissot C, Karikari TK, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27:1592.

    Article  CAS  PubMed  Google Scholar 

  188. Hoppe SC, Lin Y, Oakley D, Roe AD, DeVos SL, Hanlon D, Hyman BT. The role of microglia in processing and spreading of bioactive tau seeds in Alzheimer’s disease. J Neuroinflammation. 2018;15:15.

    Google Scholar 

  189. Majerova P, Zilkova M, Kazmerova Z, Kovac A, Paholikova K, Kovacech B, Zilka N, Novak M. Microglia display modest phagocytic capacity for extracellular tau oligomers. J Neuroinflammation. 2014;11:12.

    Article  Google Scholar 

  190. Brelstaff JH, Mason M, Katsinelos T, McEwan WA, Ghetti B, Tolkovsky AM, Spillantini MG. Microglia become hypofunctional and release metalloproteases and tau seeds when phagocytosing live neurons with P301S tau aggregates. Sci Adv. 2021;7:eabg4980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Castro-Sanchez S, Garcia-Yague AJ, Kugler S, Lastres-Becker I. CX3CR1-deficient microglia shows impaired signalling of the transcription factor NRF2: Implications in tauopathies. Redox Biol. 2019;22:12.

    Article  Google Scholar 

  192. Jin MH, Shiwaku H, Tanaka H, Obita T, Ohuchi S, Yoshioka Y, Jin XC, Kondo K, Fujita K, Homma H, et al. Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nat Commun. 2021;12:22.

    Article  Google Scholar 

  193. Wang C, Fan L, Khawaja RR, Liu BY, Zhan LH, Kodama L, Chin M, Li YQ, Le D, Zhou YG, et al. Microglial NF-kappa B drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun. 2022;13:19.

    Google Scholar 

  194. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, et al. A Unique microglia type associated with restricting development of Alzheimer’s disease. Cell. 2017;169(1276–1290): e1217.

    Google Scholar 

  195. Ruan Z, Delpech JC, Venkatesan Kalavai S, Van Enoo AA, Hu J, Ikezu S, Ikezu T. P2RX7 inhibitor suppresses exosome secretion and disease phenotype in P301S tau transgenic mice. Mol Neurodegener. 2020;15:47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Zhu B, Liu Y, Hwang S, Archuleta K, Huang H, Campos A, Murad R, Pina-Crespo J, Xu H, Huang TY. Trem2 deletion enhances tau dispersion and pathology through microglia exosomes. Mol Neurodegener. 2022;17:58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Kovacs GG. Astroglia and tau: new perspectives. Front Aging Neurosci. 2020;12:96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Sanchez-Mico MV, Jimenez S, Gomez-Arboledas A, Munoz-Castro C, Romero-Molina C, Navarro V, Sanchez-Mejias E, Nunez-Diaz C, Sanchez-Varo R, Galea E, et al. Amyloid-beta impairs the phagocytosis of dystrophic synapses by astrocytes in Alzheimer’s disease. Glia. 2021;69:997–1011.

    Article  CAS  PubMed  Google Scholar 

  199. Gomez-Arboledas A, Davila JC, Sanchez-Mejias E, Navarro V, Nunez-Diaz C, Sanchez-Varo R, Sanchez-Mico MV, Trujillo-Estrada L, Fernandez-Valenzuela JJ, Vizuete M, et al. Phagocytic clearance of presynaptic dystrophies by reactive astrocytes in Alzheimer’s disease. Glia. 2018;66:637–53.

    Article  PubMed  Google Scholar 

  200. Holmes BB, Devos SL, Kfoury N, Li M, Jacks R, Yanamandra K, Ouidja MO, Brodsky FM, Marasa J, Bagchi DP, et al. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. Proc Natl Acad Sci USA. 2013;110:E3138–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Martini-Stoica H, Cole AL, Swartzlander DB, Chen F, Wan YW, Bajaj L, Bader DA, Lee VMY, Trojanowski JQ, Liu ZD, et al. TFEB enhances astroglial uptake of extracellular tau species and reduces tau spreading. J Exp Med. 2018;215:2355–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Perea JR, Lopez E, Diez-Ballesteros JC, Avila J, Hernandez F, Bolos M. Extracellular monomeric tau is internalized by astrocytes. Front Neurosci. 2019;13:442.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Wang P, Ye YH. Filamentous recombinant human Tau activates primary astrocytes via an integrin receptor complex. Nat Commun. 2021;12:95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Chiarini A, Armato U, Gardenal E, Gui L, Dal Pra I. Amyloid beta-exposed human astrocytes overproduce phospho-tau and overrelease it within exosomes, effects suppressed by calcilytic NPS 2143-further implications for Alzheimer’s therapy. Front Neurosci. 2017;11:217.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Dinkins MB, Dasgupta S, Wang GH, Zhu G, Bieberich E. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging. 2014;35:1792–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Sharma K. Cholinesterase inhibitors as Alzheimer’s therapeutics. Mol Med Rep. 2019;20:1479–87.

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Kumar A, Singh A. Ekavali: a review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep. 2015;67:195–203.

    Article  CAS  PubMed  Google Scholar 

  208. Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, He F, Sun XY, Thomas RG, et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med. 2013;369:341–50.

    Article  CAS  PubMed  Google Scholar 

  209. Jeremic D, Jimenez-Diaz L, Navarro-Lopez JD. Past, present and future of therapeutic strategies against amyloid-beta peptides in Alzheimer’s disease: a systematic review. Ageing Res Rev. 2021;72:36.

    Article  Google Scholar 

  210. Salloway S, Chalkias S, Barkhof F, Burkett P, Barakos J, Purcell D, Suhy J, Forrestal F, Tian Y, Umans K, et al. Amyloid-related imaging abnormalities in 2 phase 3 studies evaluating aducanumab in patients with early Alzheimer disease. JAMA Neurol. 2022;79:13–21.

    Article  PubMed  Google Scholar 

  211. Mahase E. Aducanumab: European agency rejects Alzheimer’s drug over efficacy and safety concerns. BMJ. 2021;375:1.

    Google Scholar 

  212. Cummings J, Lee G, Nahed P, Kambar M, Zhong K, Fonseca J, Taghva K. Alzheimer’s disease drug development pipeline: 2022. Alzheimers Dement. 2022;8:24.

    Google Scholar 

  213. Matsunaga S, Fujishiro H, Takechi H. Efficacy and safety of glycogen synthase kinase 3 inhibitors for Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2019;69:1031–9.

    Article  CAS  PubMed  Google Scholar 

  214. Forlenza OV, Radanovic M, Talib LL, Gattaz WF. Clinical and biological effects of long-term lithium treatment in older adults with amnestic mild cognitive impairment: randomised clinical trial. Br J Psychiatry. 2019;215:668–74.

    Article  PubMed  Google Scholar 

  215. Aprahamian I, Santos FS, dos Santos B, Talib L, Diniz BS, Radanovic M, Gattaz WF, Forlenza OV. Long-term, low-dose lithium treatment does not impair renal function in the elderly: a 2-year randomized, placebo-controlled trial followed by single-blind extension. J Clin Psychiatry. 2014;75:e672-678.

    Article  CAS  PubMed  Google Scholar 

  216. Sereno L, Coma M, Rodriguez M, Sanchez-Ferrer P, Sanchez MB, Gich I, Agullo JM, Perez M, Avila J, Guardia-Laguarta C, et al. A novel GSK-3beta inhibitor reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol Dis. 2009;35:359–67.

    Article  CAS  PubMed  Google Scholar 

  217. Wang H, Huang S, Yan K, Fang X, Abussaud A, Martinez A, Sun HS, Feng ZP. Tideglusib, a chemical inhibitor of GSK3beta, attenuates hypoxic-ischemic brain injury in neonatal mice. Biochim Biophys Acta. 2016;1860:2076–85.

    Article  CAS  PubMed  Google Scholar 

  218. Lovestone S, Boada M, Dubois B, Hull M, Rinne JO, Huppertz HJ, Calero M, Andres MV, Gomez-Carrillo B, Leon T, et al. A phase II trial of tideglusib in Alzheimer’s disease. J Alzheimers Dis. 2015;45:75–88.

    Article  CAS  PubMed  Google Scholar 

  219. Hoglinger GU, Huppertz HJ, Wagenpfeil S, Andres MV, Belloch V, Leon T, Del Ser T, Investigators TM. Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a randomized trial. Mov Disord. 2014;29:479–87.

    Article  PubMed  Google Scholar 

  220. Hastings NB, Wang X, Song L, Butts BD, Grotz D, Hargreaves R, Fred Hess J, Hong KK, Huang CR, Hyde L, et al. Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener. 2017;12:39.

    Article  PubMed  PubMed Central  Google Scholar 

  221. Permanne B, Sand A, Ousson S, Neny M, Hantson J, Schubert R, Wiessner C, Quattropani A, Beher D. O-GlcNAcase inhibitor ASN90 is a multimodal drug candidate for tau and alpha-synuclein proteinopathies. ACS Chem Neurosci. 2022;13:1296–314.

    Article  CAS  PubMed  Google Scholar 

  222. Lowe SL, Goldsmith P, Phipps KM, Kevin DB, Biglan K, Mancini M, Nuthall HN, Mergott DJ, Kielbasa W. Single and multiple ascending dose studies in healthy volunteers to assess the safety and PK of LY3372689, an inhibitor of the O-GlcNAcase (OGA) enzyme. Alzheimers Dement. 2021;17: e057728.

    Article  Google Scholar 

  223. RESEARCH & DEVELOPMENT Pipeline https://www.asceneuron.com/pipeline. Accessed 10 Apr 2023.

  224. Novak P, Zilka N, Zilkova M, Kovacech B, Skrabana R, Ondrus M, Fialova L, Kontsekova E, Otto M, Novak M. AADvac1, an active immunotherapy for Alzheimer’s disease and non Alzheimer tauopathies: an overview of preclinical and clinical development. JPAD. 2019;6:63–9.

    CAS  PubMed  Google Scholar 

  225. Imbimbo BP, Ippati S, Watling M, Balducci C. A critical appraisal of tau-targeting therapies for primary and secondary tauopathies. Alzheimers Dement. 2022;18:1008–37.

    Article  CAS  PubMed  Google Scholar 

  226. Novak P, Schmidt R, Kontsekova E, Zilka N, Kovacech B, Skrabana R, Vince-Kazmerova Z, Katina S, Fialova L, Prcina M, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer’s disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol. 2017;16:123–34.

    Article  CAS  PubMed  Google Scholar 

  227. Novak P, Kovacech B, Katina S, Schmidt R, Scheltens P, Kontsekova E, Ropele S, Fialova L, Kramberger M, Paulenka-Ivanovova N, et al. ADAMANT: a placebo-controlled randomized phase 2 study of AADvac1, an active immunotherapy against pathological tau in Alzheimer’s disease. Nature aging. 2021;1:521–34.

    Article  PubMed  Google Scholar 

  228. Ji CY, Sigurdsson EM. Current status of clinical trials on tau immunotherapies. Drugs. 2021;81:1135–52.

    Article  PubMed  PubMed Central  Google Scholar 

  229. Sopko R, Golonzhka O, Arndt J, Quan C, Czerkowicz J, Cameron A, Smith B, Murugesan Y, Gibbons G, Kim SJ, et al. Characterization of tau binding by gosuranemab. Neurobiol Dis. 2020;146:18.

    Article  Google Scholar 

  230. Hoglinger GU, Litvan I, Mendonca N, Wang DL, Zheng H, Rendenbach-Mueller B, Lon HK, Jin ZY, Fisseha N, Budur K, et al. Safety and efficacy of tilavonemab in progressive supranuclear palsy: a phase 2, randomised, placebo-controlled trial. Lancet Neurol. 2021;20:182–92.

    Article  PubMed  Google Scholar 

  231. Zagotenemab | ALZFORUM, (n.d.). https://www.alzforum.org/therapeutics/zagotenemab. Accessed 10 Apr 2023.

  232. Teng E, Manser PT, Pickthorn K, Brunstein F, Blendstrup M, Sanabria Bohorquez S, Wildsmith KR, Toth B, Dolton M, Ramakrishnan V, et al. Safety and Efficacy of Semorinemab in Individuals With Prodromal to Mild Alzheimer Disease: A Randomized Clinical Trial. JAMA Neurol. 2022;79:758–67.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Congdon EE, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease. Nat Rev Neurol. 2018;14:399–415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Czerkowicz J, Chen W, Wang Q, Shen C, Wager C, Stone I, Stebbins C, Lamb M, Setser J, Cantone G, Graham D. [P4–039]: Pan-tau antibody biib076 exhibits promising safety and biomarker profile in cynomolgus monkey toxicity study. Alzheimers Dement. 2017;13:P1271–P1271.

    Article  Google Scholar 

  235. Horie K, Barthelemy NR, Sato C, Bateman RJ. CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer’s disease. Brain. 2021;144:515–27.

    Article  PubMed  Google Scholar 

  236. Panza F, Lozupone M. The challenges of anti-tau therapeutics in Alzheimer disease. Nat Rev Neurol. 2022;18:577–8.

    Article  PubMed  Google Scholar 

  237. Roberts M, Sevastou I, Imaizumi Y, Mistry K, Talma S, Dey M, Gartlon J, Ochiai H, Zhou Z, Akasofu S, et al. Pre-clinical characterisation of E2814, a high-affinity antibody targeting the microtubule-binding repeat domain of tau for passive immunotherapy in Alzheimer’s disease. Acta Neuropathol Commun. 2020;8:24.

    Article  Google Scholar 

  238. Galpern WR, Mercken M, Van Kolen K, Timmers M, Haeverans K, Janssens L, Triana-Baltzer G, Kolb HC, Jacobs T, Nandy P, et al. P1–052: A Single Ascending Dose Study To Evaluate The Safety, Tolerability, Pharmacokinetics, And Pharmacodynamics Of The Anti-Phospho-Tau Antibody Jnj-63733657 In Healthy Subjects. Alzheimers Dement. 2019;15:P252–3.

    Article  Google Scholar 

  239. Rosenqvist N, Asuni AA, Andersson CR, Christensen S, Daechsel JA, Egebjerg J, Falsig J, Helboe L, Jul P, Kartberg F, et al. Highly specific and selective anti-pS396-tau antibody C10.2 targets seeding-competent tau. Alzheimers Dement. 2018;4:521–34.

    Article  Google Scholar 

  240. Albert M, Mairet-Coello G, Danis C, Lieger S, Caillierez R, Carrier S, Skrobala E, Landrieu I, Michel A, Schmitt M, et al. Prevention of tau seeding and propagation by immunotherapy with a central tau epitope antibody. Brain. 2019;142:1736–50.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Tai H-C, Ma H-T, Huang S-C, Wu M-F, Wu C-L, Lai Y-T, Li Z-L, Margolin R, Intorcia AJ, Serrano GE, et al. The tau oligomer antibody APNmAb005 detects early-stage pathological tau enriched at synapses and rescues neuronal loss in long-term treatments. BioRxiv. 2022. https://doi.org/10.1101/2022.06.24.497452v1.

    Article  PubMed  PubMed Central  Google Scholar 

  242. Andersson CR, Falsig J, Stavenhagen JB, Christensen S, Kartberg F, Rosenqvist N, Finsen B, Pedersen JT. Antibody-mediated clearance of tau in primary mouse microglial cultures requires Fcgamma-receptor binding and functional lysosomes. Sci Rep. 2019;9:4658.

    Article  PubMed  PubMed Central  Google Scholar 

  243. Prins ND, Harrison JE, Chu HM, Blackburn K, Alam JJ, Scheltens P. Investigators R-SS: a phase 2 double-blind placebo-controlled 24-week treatment clinical study of the p38 alpha kinase inhibitor neflamapimod in mild Alzheimer’s disease. Alzheimers Res Ther. 2021;13:106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Jiang Y, Alam JJ, Gomperts SN, Maruff P, Lemstra AW, Germann UA, Stavrides PH, Darji S, Malampati S, Peddy J, et al. Preclinical and randomized clinical evaluation of the p38 alpha kinase inhibitor neflamapimod for basal forebrain cholinergic degeneration. Nat Commun. 2022;13:5308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Roy SM, Minasov G, Arancio O, Chico LW, Van Eldik LJ, Anderson WF, Pelletier JC, Watterson DM. A Selective and brain penetrant p38alphaMAPK inhibitor candidate for neurologic and neuropsychiatric disorders that attenuates neuroinflammation and cognitive dysfunction. J Med Chem. 2019;62:5298–311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Wing LK, Behanna HA, Van Eldik LJ, Watterson DM, Ralay Ranaivo H. De novo and molecular target-independent discovery of orally bioavailable lead compounds for neurological disorders. Curr Alzheimer Res. 2006;3:205–14.

    Article  CAS  PubMed  Google Scholar 

  247. Van Eldik LJ, Sawaki L, Bowen K, Laskowitz DT, Noveck RJ, Hauser B, Jordan L, Spears TG, Wu H, Watt K, et al. First-in-human studies of MW01-6-189WH, a brain-penetrant, antineuroinflammatory small-molecule drug candidate: phase 1 safety, tolerability, pharmacokinetic, and pharmacodynamic studies in healthy adult volunteers. Clin Pharmacol Drug Dev. 2021;10:131–43.

    Article  PubMed  Google Scholar 

  248. Lemos DR, Babaeijandaghi F, Low M, Chang CK, Lee ST, Fiore D, Zhang RH, Natarajan A, Nedospasov SA, Rossi FM. Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors. Nat Med. 2015;21:786–94.

    Article  CAS  PubMed  Google Scholar 

  249. Lonskaya I, Hebron ML, Desforges NM, Franjie A, Moussa CE. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med. 2013;5:1247–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Kim J, Lee HJ, Park JH, Cha BY, Hoe HS. Nilotinib modulates LPS-induced cognitive impairment and neuroinflammatory responses by regulating P38/STAT3 signaling. J Neuroinflammation. 2022;19:187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Turner RS, Hebron ML, Lawler A, Mundel EE, Yusuf N, Starr JN, Anjum M, Pagan F, Torres-Yaghi Y, Shi WK, et al. Nilotinib effects on safety, tolerability, and biomarkers in Alzheimer’s disease. Ann Neurol. 2020;88:183–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Reading CL, Ahlem CN, Murphy MF. NM101 phase III study of NE3107 in Alzheimer’s: rationale, design and therapeutic modulation of neuroinflammation and insulin resistance. Neurodegener Dis Manag. 2021;11:289–98.

    Article  PubMed  Google Scholar 

  253. Lambert WS, Carlson BJ, Formichella CR, Sappington RM, Ahlem C, Calkins DJ. Oral delivery of a synthetic sterol reduces axonopathy and inflammation in a rodent model of glaucoma. Front Neurosci. 2017;11:45.

    Article  PubMed  PubMed Central  Google Scholar 

  254. Jordan. K, Mahdavi. K, Haroon. J, Rindner. E, Zielinski. M, Venkatraman. V, Becerra. S, Goodenowe. D, Ahlem. C, Reading. C, et al: Neuroimaging data from a phase 2, open-label study of ne3107 in patients with cognitive decline due to degenerative dementias. In: Clinical Trials on Alzheimer’s Disease (CTAD) Conference; November 29–December 2; San Francisco, California, USA. 2022

  255. Callizot N, Estrella C, Burlet S, Henriques A, Brantis C, Barrier M, Campanari ML, Verwaerde P. AZP2006, a new promising treatment for Alzheimer’s and related diseases. Sci Rep. 2021;11:16806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Zhang P, Kishimoto Y, Grammatikakis I, Gottimukkala K, Cutler RG, Zhang S, Abdelmohsen K, Bohr VA, Misra Sen J, Gorospe M, Mattson MP. Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat Neurosci. 2019;22:719–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, Orr ME. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018;17: e12840.

    Article  PubMed  PubMed Central  Google Scholar 

  258. Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine. 2019;40:554–63.

    Article  PubMed  PubMed Central  Google Scholar 

  259. Gonzales MM, Garbarino VR, Marques Zilli E, Petersen RC, Kirkland JL, Tchkonia T, Musi N, Seshadri S, Craft S, Orr ME. Senolytic therapy to modulate the progression of Alzheimer’s disease (SToMP-AD): a pilot clinical trial. J Prev Alzheimers Dis. 2022;9:22–9.

    CAS  PubMed  Google Scholar 

  260. Syed YY. Sodium oligomannate: first approval. Drugs. 2020;80:441–4.

    Article  PubMed  Google Scholar 

  261. Wang XY, Sun GQ, Feng T, Zhang J, Huang X, Wang T, Xie ZQ, Chu XK, Yang J, Wang H, et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019;29:787–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Xiao SF, Chan P, Wang T, Hong Z, Wang SZ, Kuang WH, He JC, Pan XP, Zhou YY, Ji Y, et al. A 36-week multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase 3 clinical trial of sodium oligomannate for mild-to-moderate Alzheimer’s dementia. Alzheimers Res Ther. 2021;13:11.

    Article  Google Scholar 

Download references

Acknowledgements

The figure was created with BioRender.com.

Funding

This research was funded by grants from the Natural Science Foundation of Shanghai (Grant 22ZR1434700) and the National Natural Science Foundation of China (Grant 81870835).

Author information

Authors and Affiliations

Authors

Contributions

YJC and YY processed the literature and wrote the manuscript; YY conceived and designed the manuscript. All authors have read and agree with the contents of the manuscript.

Corresponding author

Correspondence to Yang Yu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing financial interests in this work.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Yu, Y. Tau and neuroinflammation in Alzheimer’s disease: interplay mechanisms and clinical translation. J Neuroinflammation 20, 165 (2023). https://doi.org/10.1186/s12974-023-02853-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-023-02853-3

Keywords