Skip to main content

Inflammation context in Alzheimer’s disease, a relationship intricate to define

Abstract

Alzheimer’s disease (AD), the most common form of dementia, is characterized by the accumulation of amyloid β (Aβ) and hyperphosphorylated tau protein aggregates. Importantly, Aβ and tau species are able to activate astrocytes and microglia, which release several proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β), together with reactive oxygen (ROS) and nitrogen species (RNS), triggering neuroinflammation. However, this inflammatory response has a dual function: it can play a protective role by increasing Aβ degradation and clearance, but it can also contribute to Aβ and tau overproduction and induce neurodegeneration and synaptic loss. Due to the significant role of inflammation in the pathogenesis of AD, several inflammatory mediators have been proposed as AD markers, such as TNF-α, IL-1β, Iba-1, GFAP, NF-κB, TLR2, and MHCII. Importantly, the use of anti-inflammatory drugs such as NSAIDs has emerged as a potential treatment against AD. Moreover, diseases related to systemic or local inflammation, including infections, cerebrovascular accidents, and obesity, have been proposed as risk factors for the development of AD. In the following review, we focus on key inflammatory processes associated with AD pathogenesis.

Introduction

Neurodegenerative diseases are a common cause of morbidity. In 2011, the worldwide cases per 10,000 inhabitants numbered 400 for Alzheimer’s disease (AD), 315 for Parkinson's disease and 30 for multiple sclerosis [1, 2]. According to the World Health Organization (WHO), approximately 50 million people have dementia, and there are nearly 10 million new cases every year worldwide. AD is the most common form of dementia and may contribute to 60–70% of cases. Most neurodegenerative diseases are related to age, thus the most vulnerable population is elderly individuals, who are more likely to suffer from AD when external factors are present, such as obesity, which exacerbates inflammation [3].

Inflammation is known to exacerbate AD pathogenesis [4]. Therefore, several research groups have proposed to treat AD with anti-inflammatory drugs. In fact, nonsteroidal anti-inflammatory drugs (NSAIDs) have been shown to delay dementia in AD [5]. NSAIDs are some of the most widely used drugs in developed countries, and their consumption continues to grow every year. In the United States, the use of NSAIDs increased by 125% from 1999 to 2002. In Spain, it was estimated that more than four million people used NSAIDs in 1999, of which 30–40% were older than 65 years. In 2005, undeveloped countries such as Costa Rica spent two million dollars on NSAIDs, which represented an increase of 25% from the expenditure in 2001 [6]. Despite advances in drugs, inflammation has not been eradicated in neurodegenerative diseases, and research on how to counteract its effects is ongoing. Numerous studies using different animal models have resulted in vital discoveries about inflammation and neuroinflammation. For example, our lab has shown that administration of andrographolide (ANDRO), a natural compound, decreases neuroinflammation in the rodent Octodon degus, which develops AD spontaneously [7]. Another study using an AD mice model induced by Aβ1–42 injection has demonstrated that ginsenoside Rb1 (GsRb1) reduces neuroinflammation in the hippocampus [8]. Additional studies have revealed a potential mechanism of action of anti-Aβ antibodies via experiments on transgenic mice overexpressing mutant human amyloid precursor protein (PDAPP) under the control of the mini-promoter of platelet-derived growth factor (PDGF) [9].

In this review, we focus mainly on the relationship between inflammation and AD through topics ranging from neurodegeneration to the roles of highly insoluble Aβ deposits and neurofibrillary tangles as inflammatory stimuli in the brains of AD patients [10]. These topics are of particular interest due to the effects of misfolded and aggregated proteins; when these proteins bind to pattern recognition receptors in microglia and astroglia, they trigger an innate immune response characterized by the release of inflammatory mediators. Accumulation of these mediators culminates in chronic neuroinflammation that exacerbates AD pathogenesis [4, 10]. External factors, including systemic inflammation, such as that observed in obesity, are likely to interfere with the brain's immune processes and further promote disease progression. Modulation of risk factors and targeting of these immune mechanisms could lead to future therapeutic or preventive strategies for AD [4].

Inflammation and neuroinflammation

To elucidate the relationship between inflammation and AD, it is necessary to understand the basics of inflammation, discuss the connection to age-related diseases, and then describe how neuroinflammation works. Inflammation is a response of the immune system to infections, tissue damage, or other harmful conditions [11, 12]. At the cellular level, the cells responsible for inflammation are immune cells such as macrophages, dendritic cells, mast cells, neutrophils, and lymphocytes, and nonimmune cells such as epithelial cells, endothelial cells, and fibroblasts. The molecules responsible for inflammation can be generally divided into cytokines and transcription factors [13].

It is important to differentiate acute inflammation from chronic inflammation. Acute inflammation is the first reaction of innate immunity and corresponds to an adaptive response, which restores cell and tissue homeostasis. The inflammatory response typically ends once the thread of damage stops, for example, when tissue repair is prepared to commence or when the pathogen is removed. This process is called the resolution of inflammation and is mediated by several anti-inflammatory mediators, such as interleukin 10 (IL-10), transforming growth factor beta (TGF-β), and glucocorticoids [13]. If this process fails, the consequences are harmful and can affect several physiological functions, such as cardiovascular, respiratory, and neuronal [13,14,15]. In this case, acute inflammation transforms into chronic inflammation. The inflammatory pathways vary with the stimulus, and the duration of the inflammatory responses changes with the level of damage. However, in most cases, these responses cause systemic effects due to the excessive production of inflammatory cytokines, leading to chronic inflammation. Chronic inflammation is the key driver of pathogenesis in several diseases, and the main damage caused to the host is mediated by the host's own inflammatory response rather than by pathogens [13]. Today, chronic inflammation is known to play a role in many diseases, such as atherosclerosis, obesity, type 2 diabetes, asthma, inflammatory bowel diseases, rheumatoid arthritis, cancer and neurodegenerative diseases [3].

Most age-related diseases, such as cardiovascular disease, cancer, metabolic syndrome, osteoporosis, dementia and AD, have an inflammatory pathogenesis [16]. In neurodegenerative diseases, inflammation has its own unique process that may present in two ways. In peripheral inflammation, cytokines such as TNF-α, interleukin 6 (IL-6) and interleukin 1β (IL-1β) can affect and cross the blood–brain barrier (BBB), stimulating it to release proinflammatory mediators and making it more permeable to cells, thus allowing the passage of leukocytes to the brain [17]. These phenomena cause a cascade of events within the brain leading to microglial and astroglial responses involving further production of proinflammatory mediators, ROS, RNS, among others. This mechanism is defined as neuroinflammation [18]. Neuroinflammation can occur, as mentioned, or can be triggered by the brain's own immune response, which depends on the glial cells. Neuronal lesions or highly insoluble proteins, such as Aβ, can activate these cells without leukocyte infiltration [19]. In both cases, neuroinflammation can culminate in synaptic deterioration, neuronal death, and exacerbation of various brain pathologies [20]. It is also important to mention that at the cellular level, astrocytes, microglia, neurons, and infiltrated leukocytes are responsible for neuroinflammation [18].

CNS cells affected by neuroinflammation in AD

The brain's innate immune system is made up mainly of microglia and astrocytes, both types of glial cells [21]. Microglia, the only immune-derived cells within the brain, are responsible for immune surveillance and response processes, including phagocytosis. On the other hand, astrocytes fulfill several homeostatic functions in the central nervous system (CNS), regulate immune responses by releasing cytokines, and act as antigen-presenting cells [22, 23]. During the chronic phase of neuroinflammation and early AD pathogenesis, the activation of microglia and astrocytes appears to play a positive effect, contributing to Aβ elimination. However, as the disease progresses, continuous activation of microglia and astrocytes in the brain can promote AD pathology [19, 24]. Indeed, sustained glial responses have been shown to correlate with Aβ plaque burden, dystrophic neurite growth, and phosphorylation of tau [25, 26].

Microglia as CNS cells affected by neuroinflammation in AD

In healthy brains, microglia are in an inactive or resting state and are distinguished both by their branched morphology and by their low expression of major histocompatibility complex (MHC) proteins and other antigen-presenting surface receptors [27, 28]. However, when extracellular signals, such as pathogens, foreign materials (such as Aβ), or dead cells, are present in the CNS, microglia undergo a morphological change to an amoeboid form, regarded as “activated” microglia [24]. As a consequence, cytokines and proinflammatory mediators, such as IL-1β, IL-6, and TNF-α, and ROS are released, leading to neuronal damage [19]. This uncontrolled and sustained activation of the microglia state is described as reactive gliosis and has been implicated in AD pathogenesis [29]. Interestingly, several studies have shown that activated microglia are predominantly found in close vicinity to Aβ plaques [30,31,32]. Similarly, the formation and appearance of Aβ plaques in the brain correlate with the activation of microglia [33]. As a consequence, this microglia accumulation can worsen AD pathology and neurodegenerative processes by overexpressing proinflammatory cytokines, which in turn may lead to a reduction in Aβ clearance and accumulation in the brain [24]. For example, interferon γ (IFNγ) and TNF-α, two chemokines released by activated microglia, have been demonstrated to decrease Aβ degradation and increase its production [34, 35]. Contrarily, genetic deletion of IFNγ receptor was shown to reduce microglial activation and Aβ generation, together with the prevention of cognitive decline [34, 36].

Astrocytes as CNS cells affected by neuroinflammation in AD

Astrocytes are CNS resident cells and play several key roles in brain homeostasis maintenance, including neurotransmitter uptake, blood flow regulation, and support of neuronal metabolism, among others [37]. However, as a response to many pathological situations, including trauma, neuroinflammation, or neurodegeneration, these homeostatic astrocytes change to a reactive phenotype and induce astrogliosis [38]. The latter is described as an abnormal increase of astrocytes, and it is characterized by cellular hypertrophy and an increase in the glial fibrillary acidic protein (GFAP) expression [39]. Moreover, activated astrocytes exert neurotoxic effects with loss of neurotrophic functions, causing a chain reaction. These events are associated with increased release of cytokines and inflammatory mediators, neurodegeneration, decreased uptake of glutamate, and loss of neuronal synapses [24]. In AD, astrogliosis can be triggered both by damaged neurons or glia and by extracellular deposits of Aβ [40,41,42]. Indeed, Aβ can induce signals and spontaneous oscillations of intracellular calcium concentrations in astrocytes that are partially responsible for neurotoxicity [43]. In addition, studies in cultures have shown that Aβ decreases the expression and capacity of two astrocytic glutamate transporters, glutamate-aspartate transporters, and glutamate transporter 1, which culminates in a decrease in glutamate uptake by astrocytes, inducing excitotoxicity [43, 44]. Another characteristic of astrocytes that defines them as detrimental in the pathogenesis of AD is their release of proinflammatory cytokines and ROS, which may also lead to neuroinflammation and neurotoxicity [45].

Neuroinflammation related to Aβ and tau

The presence of aggregated Aβ and tau are hallmark pathogenic features in AD and can be found in the early and late stages of the disease. Importantly, inflammation can trigger Aβ or tau overproduction, which in turn induces inflammatory responses, resulting in a vicious cycle of neuroinflammation and pathology [46,47,48].

Neuroinflammation related to Aβ

The appearance of amyloid plaques, which are primarily composed of Aβ, is accompanied by strong activation of microglia in the vicinity of the plaques, suggesting that Aβ acts as one of the primary drivers of microglial activation. Indeed, injecting Aβ directly into the brain has been shown to induce microglial activation and neuronal loss [49]. However, studies using transgenic mice overexpressing amyloid precursor protein (APP) have revealed contradictory effects on AD pathogenesis depending on the microglial phenotype [50]. For example, M1 microglia (classically activated) produce neurotoxic proinflammatory mediators, including cytokines (e.g., TNF-α and IL-1β), chemokines (e.g., CCL2), and ROS/RNS. Overall, this neuroinflammatory response leads to neuronal damage. In addition, the microglial clearance mechanisms of Aβ participate in localized inflammatory mechanisms that can be cytotoxic to nearby tissue [51]. On the other hand, during M2 microglia phenotype (alternative macrophage activation), phagocytosis of Aβ occurs, and anti-inflammatory and neurotrophic mediators are expressed and produced, such as arginase 1, BDNF, IGF-1, and IL-10. These molecules provide protection to neurons and have been associated with neuronal regeneration (Fig. 1) [52]. It is important to mention that the microglial activation phenotype may change during AD. In the early stage of the disease, M2 microglia predominate to engulf the Aβ and produce anti-inflammatory factors to quench proinflammation and maintain tissue homeostasis. However, the persistent presence of Aβ plaques promotes polarization of microglia toward the M1 phenotype, thus compromising the immune resolution process in the later stage of disease progression and leading to neuronal degeneration (Fig. 1) [53].

Fig. 1
figure 1

Microglial M2 and M1 phenotypes during AD. Two microglial activation phenotypes fluctuate in AD: M1, the classic and inflammatory phenotype (associated with release of ROS, RNS, and proinflammatory cytokines such as TNF-α and IL-1β); and M2, the alternative and anti-inflammatory phenotype (associated with release of anti-inflammatory cytokines such as IL10 and IGF1), which is responsible for Aβ phagocytosis. M1-to-M2 polarization is stimulated by proinflammatory cytokines such as IL4 and IL13, while M2-to-M1 polarization is stimulated by a lack of anti-inflammatory cytokines such as IL10 and IGF1. The M1 phenotype is seen in earlier stages of the disease, while the M2 phenotype is seen later. An example of a receptor involved in M2 activation is the nuclear receptor PPARγ, and an example of a pathway involved in M2 activation is the LKB1-AMPK pathway. An example of a receptor involved in M2 activation is the TLR2 receptor, which is capable of binding to Aβ, and an example of a pathway involved in M2 activation is the MYD88 pathway

Due to the action of Toll-like receptors (TLRs) present in microglia, Aβ ultimately causes neuroinflammation [54]. For example, one study found that TLR2 interacts directly with Aβ42 [52]. In fact, when TLR2 is expressed in HEK293 cells that do not express TLR2 endogenously, the cells respond to Aβ42 and release IL-8 (a proinflammatory cytokine). The leucine-rich repeats (LRRs) in the N-terminal domain of the TLR2 receptor are partly responsible for this ligand-receptor interaction, but not completely, because TLR1 and TLR3 also contain LRRs and do not bind to Aβ. Another important finding from the same study was the identification, by directed mutagenesis, of the amino acids EKKA (741–744) as a critical cytoplasmic domain for inflammatory signal transduction. The results also showed that the TLR-MyD88 signaling pathway controls M1 microglial activation. In fact, the authors observed that myeloid differentiation primary response 88 (MyD88) deficiency increases Aβ phagocytosis but decreases inflammatory activation. These findings suggest that the signaling pathway that controls M1 microglial activation in response to Aβ is separated from the signaling pathway that activates M2 microglia in response to Aβ and increases phagocytosis of Aβ. Furthermore, the authors observed that in transgenic mouse-derived bone marrow (BM) precursors with chimeric AD amyloid, that is, TLR2-deficient BM-derived microglia, more M2 microglia were activated than M1 microglia, which reduces neuroinflammation. In summary, Aβ binds to TLR2, causing M1 microglial activation, and signal transduction through the MyD88 pathway ends with the release of proinflammatory mediators that induce neuroinflammation. This process contributes to the dysfunction and neuronal loss characteristic of the pathogenesis of AD (Fig. 2) [50, 52].

Fig. 2
figure 2

Vicious cycle of neuroinflammation with Aβ and tau. Aβ and tau activate microglia and astrocytes to produce ROS, RNS, and proinflammatory cytokines such as IL-1 and TNF-α, inducing neuroinflammation. In turn, neuroinflammation increases the levels of Aβ and tau (because it is involved in the hyperphosphorylation of tau and attenuates the clearance of Aβ) or causes neurodegeneration in different ways (for example, via the PAP-2, GMF, ROS and RNS pathways). Neurodegeneration exacerbates neuroinflammation because the remains of dead cells and the tau that remains in the extracellular environment due to cell death activate microglia and astrocytes. This increased neuroinflammation also increases the Aβ and tau levels (for the same reasons as in the initial neuroinflammation)

Efforts have been made to study the relationship among signaling pathways, microglia, and AD. Indeed, recent evidence has shown that adenosine monophosphate-activated kinase (AMPK) signaling is involved in microglial polarization [55]. Also, peroxisome proliferator-activated receptor γ (PPARγ) has been shown to regulate the M2 microglial activation (Fig. 2). In fact, in one study, increased mRNA levels of the M2 marker YM1 were observed when cells were treated with PPARγ agonists [56]. In addition, several studies have revealed that treatment with agonists of PPARγ reduces Aβ levels in the CNS and alleviates AD pathology [56,57,58]. The modulating effect of PPARγ on microglial polarization could be due to activation of the LKB1-AMPK signaling pathway; this is inferred because in one experiment, the results indicated that treatment with the LKB1 inhibitor radicicol or removal of LKB1 prevented the activation of AMPK signaling and the phenotypic shift from M1 to M2 in the BV2 microglial cell line treated with LPS. This information could be used for future AD treatment studies [59, 60].

Although gliosis has been mostly shown to occur as a consequence of amyloid plaques, early evidence suggests that Aβ and tau oligomeric species can also activate the immune system. Indeed, several studies have shown the activation of different microglial and astrocytic markers and the release of proinflammatory molecules prior to the deposition of Aβ plaques [61,62,63,64]. More importantly, the inflammatory response appears to correlate with the concentration of oligomers but not to the plaque burden or monomeric Aβ. Though it is not yet entirely understood how these toxic oligomers perform their activity [63], they have been associated with several microglial and astrocytic membrane proteins. Many of these receptors, including N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, cause a fast influx of Ca2+, triggering inflammation[65,66,67]. In addition, Aβ can also bind the receptor for advanced glycation end products (RAGE), a surface molecule found in microglia, astrocytes, and cerebral endothelial cells, which mediates immune responses [68]. Interestingly, Aβ oligomers have been shown to significantly upregulate RAGE levels [69,70,71], however, it remains controversial which Aβ species induces inflammation[72].

Neuroinflammation related to tau

In addition to the presence of Aβ plaques, another characteristic pathogenic feature of AD is the presence of intraneuronal neurofibrillary tangles (NFTs) that consist of aggregated and abnormally hyperphosphorylated tau protein [73]. What triggers the formation of paired helical filaments is currently unknown, however, neuroinflammation could play a role since it is involved in hyperphosphorylation of tau and the formation of NFTs [74]. In turn, the presence of NFT exacerbate neuroinflammation and neurodegeneration by binding to microglia and activating them to produce neuroinflammatory mediators [75]. In fact, in a study using P301S-mutant human tau-transgenic mice, it was observed that microglial activation preceded the formation of tangles, and immunosuppression in these mice decreased tau pathology and increased life expectancy [74].

The hyperphosphorylated tau protein ultimately causes neurodegeneration in different ways ranging from microtubule destabilization to interaction with the 20S subunit of the proteasome and inhibition of its activity, which contributes to abnormal accumulation of proteins and initiates a cascade of events that leads to neuronal death [75, 76]. Once neuronal death has occurred, hyperphosphorylated tau aggregates are released into the extracellular environment, activating glia, which may lead to neuroinflammation [76]. Although different transgenic models of tauopathy have been used, age-dependent microglial activation-related astrogliosis and neuroinflammation have been observed at a stage in which neuronal loss does not occur [75]. It has been shown that treatment with extracellular tau activates p38 MAPK and alters the proinflammatory cytokine expression pathway in microglia, increasing the gene expression of IL-6, IL-1β, TNF-α and Mip-1α [77]. In addition, microglia activation has been shown to occur through the chemokine receptor CX3CR1 and its ligand fractalkine (CX3CL1) [78]. Under physiological conditions, CX3CR1/ CX3CL1 signaling is involved in the synthesis of anti-inflammatory signals, however, in AD, the expression of CX3CL1 was shown to be decreased. Importantly, tau has been shown to compete with CX3CL1 by binding and triggering the internalization of CX3CR1 [79], which can accelerate tau pathology and memory impairment [73, 80].

Similar to Aβ, tau oligomers have also been shown to initiate an inflammatory response [81]. Recent evidence demonstrated that tau oligomers promote the release of high mobility group box 1 (HMGB1), a highly expressed protein in AD which is able to activate RAGE and TLR4, promoting the expression of inflammatory cytokines [81, 82]. Additional studies also found a tau oligomer-dependent activation of microglia; however, the mechanism remains less clear [83, 84]. The effects of both Aβ accumulation and tau hyperphosphorylation on glial activation and neuroinflammation are presented in Fig. 2.

Inflammation markers in AD

Inflammatory markers have begun to be exploited for age-related diseases. Among the inflammatory markers whose levels are increased in age-related diseases are IL-1β and TNF-α. Moreover, the transcription factors that regulate chronic inflammation in different diseases are nuclear factor kappa-activated light chain B (NF-κB), signal transducer and activator of transcription (STAT). These proteins positively regulate several genes that encode proinflammatory cytokines and can also be used as inflammatory markers [3]. Inflammatory markers are crucial in the study and diagnosis of AD. As stated earlier, inflammation participates in the development of AD and is present in CNS cells, such as microglia and astrocytes; as well as in pro-inflammatory mediators and transcription factors (Table 1).

Table 1 Descriptions of different inflammatory markers used for the study or diagnosis of AD

Microglial inflammation markers in AD

As stated earlier, microglial activation can be used as an indicator of neuroinflammation in AD, since microglia, in the presence of insoluble proteins such as those found in AD, release proinflammatory factors [35]. For this reason, microglial activation markers can be used as neuroinflammatory markers in AD. Many microglial proteins are used as markers of neuroinflammation, such as TLR2, which interacts with Aβ; MHC II, a protein present in activated microglia that presents digested and phagocytosed fragments of pathogens, increasing the inflammatory response in surrounding microglial cells; and IBA-1, an intracellular calcium-binding protein related to the reorganization and support of the phagocytosis process because of its ability to bind to actin molecules [74, 76, 85].

In addition, genetic factors such as rare variants of the triggering receptor expressed on myeloid cells 2 (Trem2) have been recently suggested to play a crucial role in AD pathogenesis [86]. Trem2 is a type I transmembrane protein which recognizes phospholipids, apoptotic cells and lipoproteins, among others [87,88,89,90]. Studies in AD mouse models have suggested that Trem2-dependent activation of microglia is necessary to limit Aβ pathology [91]. Indeed, Trem2 is one of the most highly expressed receptors in microglia and can affect Aβ clearance in a manner mediated by phagocytosis of apoptotic and inflammatory neurons [90, 92, 93]. Interestingly, TREM2 knockout (KO) animals have shown attenuated cytokine production in response to Aβ [94, 95]. Although it is not clear how variants of the TREM2 gene confer a two- to fourfold increased risk for sporadic AD [96, 97], it has been postulated that Trem2 deficiency produces numerous effects. For example, it reduces the viability and proliferation of primary microglia, as microgliosis is reduced in Trem2-/- mouse brains; induces cell cycle arrest at the G1/S checkpoint; and ultimately reduces the stability of catenin, a key component of the canonical Wnt signaling pathway that is responsible for (among many other biological processes) cell survival. Thus, it has been shown that the Trem2-mediated Wnt/β-catenin pathway plays a pivotal role in microglial viability, suggesting that therapeutic modulation of this pathway can help combat the deterioration of microglial survival and microgliosis associated with AD [98]. Moreover, mutations in the TREM2 gene, such as Y38C, have been related to an increased risk of AD comparable to the risk associated with mutations in the allele of the apolipoprotein E (APOE) [96, 99, 100]. However, the effect of Trem2 on Aβ pathology has been inconsistent. Indeed, some studies using Trem2-/- mice have found no change in total plaque load [101, 102], while other studies have found an increase [89, 103] or even a decrease in load [103, 104]. Some of these differences, which may be attributable to the data collection time windows [103], could suggest that the elimination of Aβ is not the only function of Trem2 [105].

Similarly, Trem2 deficiency has been shown to both mitigate neuroinflammation and protect against brain atrophy in the context of tau pathology, as well as to accelerate tau aggregation [106,107,108]. For example, attenuation of neurodegeneration and significantly reduced microgliosis have been observed in Trem2-deficient mice crossed with the PS19 human tau transgenic line [108]. However, the authors did not see changes in tau phosphorylation or insoluble tau levels. These observations suggest that Trem2 can facilitate a microglial response in the context of tau pathology or tau-mediated damage in the brain without altering tau levels. Furthermore, there is evidence suggesting that microglia may contribute to the neurodegenerative process associated with tau pathology without altering tau aggregation [109]. On the other hand, using a humanized tau model, Bemiller and colleagues showed that Trem2 deficiency altered the microglial response and accelerated the aggregation and hyperphosphorylation of tau [106]. Likewise, the absence of Trem2 has been associated with a decrease in microgliosis in a variety of disease models, but the ultimate effect on the different pathologies and neuronal integrity differs, suggesting that Trem2 mediates microglial responses to amyloidosis even though it does not necessarily affect the total plaque load or protein aggregation [110, 111]. Together, these results suggest dual roles of Trem2 and microglia in the context of both amyloid and tau pathology.

Trem2 signals through different pathways, which can be modulated to antagonize the detrimental effects. For example, recent evidence shows that Trem2 promotes microglial survival by activating the Wnt/β-catenin signaling pathway; thus, it is possible to restore Wnt/β-catenin signaling when Trem2 activity is disrupted or reduced [98]. Similarly, studies using transgenic mouse models of AD indicate that inefficient mTOR signaling in Trem2-deficient microglia is associated with a compensatory increase in autophagy in vitro in AD [112,113,114]. Likewise, cell membrane phospholipids and lipoprotein particles can continuously activate Trem2, inducing mTOR signaling through upstream activators such as PI3-K, PDK1, and Akt, which are recruited by signaling associated with the Trem2 subunits DNAX-activating protein (DAP12) and DAP10 [114]. Additionally, other groups have reported that Trem2 deficiency alters the expression and secretion of cytokines induced by Aβ, and its downstream signaling alters the expression of proinflammatory cytokines such as IL-6 and macrophage inflammatory protein 1α (MIP-1α, also known as CCL3), and decreases the expression of the anti-inflammatory protein Arg1 [95, 106].

In addition to Trem2, several immune genes have also been implicated in AD pathology. Among them is CD33, a microglial receptor that has been shown to be elevated in AD [115, 116]. Importantly, several polymorphisms in CD33 have been associated with AD susceptibility and a correlation between CD33 expression and cognitive function has been described previously. More importantly, CD33 depletion results in a reduction of Aβ levels, together with a decrease plaque burden [117]. Another immune gene associated with AD is phospholipase C-gamma 2 (PLCG2), which is expressed primarily by microglia and granule cells [118]. Interestingly, PLCG2 expression was also shown to correlate with Aβ density [119]. A third gene recently discovered by genome-wide association studies associated with microglia and AD is inositol polyphosphate-5-phosphatase (INPP5D) [120]. Similar to CD33 and PLCG2, the expression of INPP5D was shown to correlate with AD progression and amyloid plaque density [121].

Astrocytic inflammation markers in AD

Likewise, astrocyte markers can also be used as indicators of neuroinflammation [122, 123]. Increased GFAP mRNA levels have been associated with AD since GFAP levels in AD brains are almost twice of those in control brains [39]. Furthermore, GFAP levels are strongly and positively correlated with the duration of the disease, suggesting that GFAP can be a useful neuroinflammatory marker in AD [124].

Proinflammatory cytokines as inflammation markers in AD

Other inflammatory markers in AD are proinflammatory cytokines, such as TNF-α and IL-1 [125]. Indeed, in an early study, Tan and colleagues followed cognitively healthy participants with the aim to determine whether cytokines, such as IL-1, IL-6 and TNF-α levels were associated with AD risks [126]. Interestingly, individuals with the highest production of these cytokines were at higher risk of developing AD. The inflammatory cytokine TNF-α is particularly important in the development of AD since it participates in the spread of inflammation and plays a critical role in the pathophysiology of AD [127]. Several studies have shown that TNF-α levels are significantly higher in patients with AD than in healthy people [128,129,130]. In fact, in one study, injecting mice with Aβ1–40 increased the expression of TNF-α, and worsened cognitive function [131]. Importantly, the cognitive impairment was alleviated by treatment with anti-TNF-α antibodies and led to a reduction of biochemical alterations produced by Aβ1–40.

NF-κB, a transcription factor related to AD

The last marker of inflammation that will be discussed is the transcription factor NF-κB, which regulates the transcription of several proinflammatory genes and is activated by Aβ, tau, ROS, and several cytokines, among other molecules [132, 133].

NF-κB dysregulation has been widely associated with AD and can result in glial cell activation [134]. In addition, a strong activation of NF-κB has been found in cells treated with different Aβ fragments, such as Aβ1-40 [133]. In a different study, the activity of the transcription factor NF-κB was measured using a microglial cell line treated with sAPP (the peptide remaining after cleavage of APP with alpha or beta-secretase). Interestingly, sAPP-treated cells responded with the activation of the transcription factor NF-κB, suggesting a close association between these molecules [135]. Indeed, the levels of NF-κB p65, one of the members of the NF-κB family, were found to be significantly increased in the brains of patients with AD, and two functional NF-κB binding elements were identified in the promoter region of the human BACE1 gene [136], which can further worsen AD pathology. Thus, given the association between TNF-κB and neurodegeneration/inflammation, attenuating this pathway could provide a therapeutic avenue to attenuate AD pathology [136, 137].

Similar to Aβ, a recent study found that tau caused the activation of the NF-κB pathway in microglia, which in turn led to an increase in tau seeding and spreading [138]. More importantly, the inactivation of microglial NF-κB was able to restore cognitive deficits and helped to reestablish a homeostatic phenotype in microglia.

Implications of neuroinflammation in AD

AD is largely characterized by cognitive deficits that arise due to neurodegeneration or synaptic loss [139]. In this section, we will discuss how the neuroinflammation present in AD leads to neurodegeneration and synaptic loss [140].

Neurodegeneration as an implication of neuroinflammation in AD

Neurodegeneration is a phenomenon that occurs in the CNS through signals associated with the loss of neuronal structure and function [141]. In AD, neurons in the hippocampus and entorhinal cortex are the first to degenerate [142]. Neurodegeneration is mediated, among other factors, by inflammatory and neurotoxic mediators such as IL-1β, TNF-α, ROS, and RNS [143]. These mediators directly or indirectly affect neuronal survival and induce neurodegeneration through glial cells and inflammatory cells. Activated microglia, astrocytes, neurons, T cells, and mast cells release these inflammatory mediators [141]. For example, when there is dysfunction in the BBB caused by microglia-secreted proinflammatory mediators in AD, peripheral inflammatory mediators and immune and inflammatory cells, such as T cells and mast cells, cross the BBB [144]. These mediators and cells activate microglia, astrocytes, and neurons to release even more proinflammatory mediators, increasing the inflammation-mediated neurodegeneration [141]. Several mediators, receptor proteins, or pathways are upregulated or activated in neuroinflammation and mediate neurodegeneration, such as protease-activated receptor 2 (PAR-2), which is expressed in mast cells, glial cells, and neurons. Mast cell activation induces the release of specific types of extracellular vesicles (EVs) with specific inflammatory mediators, including proteases such as tryptase, which activate neurons and glial cells and increase neuroinflammation mediated by the PAR-2 pathway, regulating neurodegeneration [143]. Another mediator is glial maturation factor (GMF), a protein that activates microglia and neurons, causing these cells to release proinflammatory cytokines and thus mediating neurodegeneration [143].

The last examples of neurodegeneration mediators that will be discussed in this section are ROS and RNS. Notably, microglia respond to neuroinflammation by changing their gene expression, including via de novo expression of the inducible isoform of nitric oxide synthase (iNOS), thus triggering oxidative and nitrosative stress [145]. Mitochondria are targets of oxidative and nitrosative stress, as ROS and RNS damage proteins, nucleic acids, polysaccharides, and lipids, which can lead to damage and even cause mutations in mitochondrial DNA. Overall, mitochondrial alterations can result in neurodegeneration [146]. Given the relationship between mitochondrial dysfunction and AD, treatments that target mitochondria, such as the mitochondrial antioxidant Szeto-Schiller peptides, have been widely studied [147]. These peptides have a sequence motif that allows them to target mitochondria, and their antioxidant action can be attributed to tyrosine or dimethyltyrosine (Dmt), which plays a role in mitochondrial ROS clearance. These drugs are currently undergoing phase II clinical trials for the treatment of diseases involving mitochondrial oxidative damage, such as AD and other neurodegenerative diseases [148].

Synaptic loss as an implication of neuroinflammation in AD

It has been established that neuroinflammation can lead to synaptic loss. Pro-inflammatory cytokines such as IL-1β and TNF-α regulate the transcription of many genes, including genes encoding enzymes in the cascade of arachidonic acid (a precursor of prostaglandin) in various cell types [149]. In the brain, upregulated arachidonic acid and its metabolites influence signal transduction and transcription. For example, they can influence the transcription of synaptic proteins, causing synaptic loss [150]. An example of an altered synaptic protein is the synaptic marker drebrin, whose levels are decreased in AD [151]. In fact, in one experiment, drebrin levels in the superior temporal cortex were found to be approximately 35% lower in subjects with mild cognitive impairment, who exhibited upregulation of TNF-α and IL-6, than in subjects without cognitive impairment [152].

Anti-inflammatory drugs for AD treatment

NSAIDs are widely used medications to reduce pain, fever, and other inflammatory processes and exert their anti-inflammatory function by inhibiting the enzyme cyclooxygenase (COX), which converts arachidonic acid into prostaglandins [153]. Although AD has no cure, several research lines have proposed the use of NSAIDs to alleviate symptoms [154]. NSAIDs have been used since the 2000s, and their use for AD has been tested in animal models and clinical trials [155,156,157]. Indeed, experiments in animal models of AD have shown that NSAIDs can be useful in this pathology. For example, in transgenic mice overexpressing APP, oral administration of ibuprofen, a nonspecific inhibitor of COX, at the onset of amyloid plaque formation decreased glial activation and plaque density [158, 159]. In another experiment, rats injected with Aβ in the dentate gyrus, treatment with indomethacin attenuated microglial activation, restored long-term enhancement of the hippocampus and prevented deficits in working memory. Moreover, in mice injected intracerebroventricularly with Aβ, increased COX-2 levels and memory impairment was induced [160]. Importantly, these alterations were attenuated by pretreatment with NS398, a selective COX-2 inhibitor. Additional reports have shown that ibuprofen and naproxen treatment in transgenic mouse models of AD were able to block microglial alterations without affecting APP processing [161]. There have also been studies in human cell cultures that have sparked hope regarding the treatment of AD with NSAIDs. For example, in an experiment using human neuroglioma cells overexpressing APP695NL, researchers observed that multiple NSAIDs such as sulindac, ibuprofen, and diclofenac selectively reduced Aβ42 [162]. Long-term placebo-controlled clinical trials have also demonstrated the usefulness of NSAIDs in AD. Among them is a study that evaluated the protective effects of naproxen, a nonselective COX inhibitor, and celecoxib, a selective COX-2 inhibitor, against AD in cognitively normal individuals over 70 years of age with a family history of AD [161]. Interestingly, subjects previously exposed to naproxen were 67% less likely to develop AD than control subjects previously exposed to placebo. However, it is important to note that these beneficial effects were observed only in people who had completely normal brains at the start of the study and not in people with existing brain disease (even if asymptomatic) at the start of the trial. It is worth noting that some of these people even worsened after NSAID administration [161, 163, 164]. Therefore, it can be suggested that chronic use of NSAIDs may be beneficial only in the early stages of the AD process, coinciding with the initial deposition of Aβ, the activation of microglia and the subsequent release of proinflammatory mediators. When the Aβ deposition process has already begun, NSAIDs are useless and can even be harmful, as they inhibit microglial inflammation; such inflammation, despite having harmful effects, mediates the elimination of Aβ [161]. It has been observed in other clinical trials that the protective effects of NSAIDs are superior when the duration of administration is longer than 12 months [161]. In addition, it has been suggested that the treatment time required to obtain the full benefit is two years [165].

Among the best-characterized reasons why NSAIDs can help alleviate AD symptoms are through the inhibition of COX. Research involving several animal models of AD has shown altered COX-2 brain expression, together with reactive gliosis, and behavioral dysfunction [160]. The increase in COX-2 may arise from microglial activation directly or indirectly caused by Aβ [166]. Indeed, postmortem AD brains show elevated levels of COX-1 and COX-2, compared to control brains [167]. COX-2 expression also increases after microglial activation. In fact, transgenic mice that overexpress COX-2 develop an age-dependent deficit in spatial memory at 12 and 20 months that is accompanied by apoptosis of neurons and astrocytes [160]. Interestingly, COX-2 is expressed in high concentrations in degenerative cells of the brain, thus inhibiting COX may reduce the risk of developing AD [168]. One study measured LPS-stimulated inhibition of plasma prostaglandin E2 (PGE2), an index of COX-2 activity ex vivo in volunteers who received placebo or various doses of celecoxib, a selective COX-2 inhibitor [169]. The results indicated that the COX-2 activity was significantly lower in volunteers who received different doses of celecoxib than in volunteers who received a placebo. Another reason why NSAIDs are associated with delayed AD symptoms is their effect on prostaglandins. Studies on the inflammatory mechanisms of AD have revealed that prostaglandins released during the inflammatory reaction have a degenerative effect [170]. In addition, prostaglandins can cause Aβ levels to rise. The last reason is not directly related to the anti-inflammatory capacity of NSAIDs but rather is related to the fact that these drugs reduce Aβ and tau levels. Some NSAIDs, such as indomethacin and ibuprofen, among others, have been reported to reduce the production of amyloid Aβ42 independently of COX inhibition [165]. Other hypotheses state that NSAIDs can interact directly with Aβ, thus preventing its accumulation. For example, ibuprofen and indomethacin have been shown to decrease the production of Aβ42 in vitro and in AD transgenic mice [161].

Animal models for the study of neuroinflammation in AD

The relationship between inflammation and AD discussed thus far was discovered through studies on animal models of AD. It is also very important to acquire knowledge about possible treatments or risk factors for AD, which can also be achieved through the use of animal models. Such research is essential because AD is a disease without a cure. However, animal models can be used to investigate new drugs or treatments that can improve symptoms or even cure the disease.

Several different types of animal models are used to study AD, such as transgenic mice or rat models. Transgenic animals reproduce aspects of AD, such as mice that overexpress APP or mice that express human tau isoforms with typical AD mutations [171]. An example of a transgenic mouse is the PDAPP mouse, which overexpresses APP under the control of the mini-promoter of PDGF. This mouse model shows many of the pathological characteristics of AD, such as extensive deposition of extracellular amyloid plaques, astrogliosis, and neuritic dystrophy [172, 173]. Another example is the transgenic mouse expressing a human isoform of tau that lacks the two amino-terminal inserts (since it has a P301L mutation) under the control of the murine PrP promoter [174]. These mice exhibit NFTs in the BM and brain, and in addition, 90% of these mice develop motor and behavioral disturbances by 10 months of age [173, 175]. A third animal model is O. degus, a rodent that develops AD pathology without genetic manipulation or intervention. At approximately 56 months of age, features of AD are observed, such as the expression of neuronal β-APP (β-APP695), a neural-specific isoform containing 695 amino acids, intracellular and extracellular deposition of Aβ, intracellular accumulation of tau protein and ubiquitin, a strong astrocytic response, and the appearance of markers of AD [176, 177]. In addition, AD can be studied by imitating the characteristics of AD in a non-transgenic way. Although these models are not considered AD models, they have been of great use in various findings. For example, injecting Aβ1–42 into mice induces neuronal death in the CA1 region of the hippocampus, activates astrocytes and microglia, induces the expression of nitric oxide synthase, and triggers memory loss, among other AD features [178,179,180].

Studies on animals have contributed to some major therapeutic advances. For example, experiments to demonstrate the effects of ANDRO on AD have been performed in O. degus. Indeed, one study analyzed the expression of the neuroinflammatory markers GFAP, IL-6, and COX-2. Higher levels of GFAP and IL-6 were measured in the hippocampi of old animals than in young animals. The authors also observed approximately 30% lower levels of GFAP and approximately 40% lower levels of IL-6 in the hippocampi of 56-month-old animals treated with ANDRO (injected intraperitoneally) than in those of control animals (injected with a vehicle) of the same age or in those of 12-month-old control animals. Therefore, evidence suggests that ANDRO treatment can significantly decrease neuroinflammation [7]. In another study, rats were injected intraventricularly with Aβ1–42 to emulate the presence of Aβ in AD. In that study, the authors studied the effects of GsRb1, an anti-inflammatory component of Panax ginseng and one of the most commonly used medicinal herbs in Asian and Western countries [181], on behavior and the levels of inflammatory mediators. After 2 weeks of Aβ1–42 injection their results revealed that the model rats (injected with Aβ1–42 and not treated with GsRb1) showed a significant increase in the time taken to find the hidden platform. No loss of locomotor performance was observed, suggesting that the increase in time was due to a slowed learning process. After 4 weeks of treatment with GsRb1, learning ability was restored in the treated group, as indicated by a decrease in the platform encounter time compared to that of the model group. Moreover, the number of COX-2-immunopositive cells within the hippocampus was significantly greater in the model rats than in the treated rats, indicating that GsRb1 decreased COX-2 expression and thus regulated behavior. Similar results were obtained for IkB-α, and for nNOS: the immunopositive cells were greater in number in the treated rats than in the model rats. Through the use of the Aβ1-42 injection model, the authors concluded that GsRb1 reversed changes in various markers of neuroinflammation in the hippocampus, suggesting that this anti-inflammatory agent can be used to develop antiaging drugs [8].

Finally, we will discuss a study in which the authors proposed that certain anti-Aβ antibodies exert their effects through an antibody-mediated microglial activation [9]. In that study, the researchers administered an anti-Aβ antibody (m3D6) that binds aggregated Aβ in a PDAPP transgenic mouse model, which was further engineered to contain green fluorescent protein (GFP)-expressing fluorescent microglia. Interestingly, animals injected with m3D6 had significantly more microglial cells and had almost twice the number of processes protruding from their cell bodies [9].

Obesity and AD

As discussed throughout this review, inflammation plays an important role in the pathogenesis of AD. Therefore, pathologies related to inflammation, such as stroke, traumatic brain injury, type 2 diabetes, metabolic syndrome, infection, sepsis, and obesity, are risk factors for AD [182,183,184,185,186]. Most cases of AD, approximately 95%, correspond to sporadic and nonfamilial AD, and thus it is important to understand the risk factors associated with this disease to help prevent it [187]. Among these factors is obesity, which has been proposed to influence AD progression [188]. According to the WHO, in 2016, approximately 40% of adults over 18 were overweight, and 13% were obese, while more than 340 million children and adolescents were overweight or obese [189]. Importantly, obesity has been shown to induce a state of inflammation [190]. For example, in one study, researchers analyzed the levels of the proinflammatory cytokine TNF-α in blood samples of obese and non-obese people ranging from 20 to 60 years old. Their findings showed that the TNF-α serum levels were significantly higher in obese individuals than in non-obese individuals [190]. In a different study, Hahm and colleagues evaluated the effect of a high-fat diet (HFD) on Aβ deposition and neuroinflammation [191]. Interestingly, their findings revealed that the HFD group exhibited a significant increase of approximately twofold in oxidative stress, measured by ROS and lipid peroxidation. This increase in oxidative stress also led to higher insulin resistance by impairing the adiponectin receptor 1 (AdipoR1) mediated AMPK-activated protein kinase (AMPK) signaling, which can exacerbate the pathogenesis of AD. Similarly, the suppression of AdipoR1 also resulted in increased oxidative stress and increased amyloidogenic pathway. Finally, the researchers observed that HFD feeding increased the levels of Aβ by more than 50% in the cortex and hippocampus (Fig. 3). Together, these results suggest that obesity, through inflammation, influences AD pathogenesis. Moreover, decreasing obesity, eating a healthy diet, and performing physical activity could be useful habits to prevent AD [192, 193].

Fig. 3
figure 3

Brain inflammation produced in obesity. In HFD-fed mice, peripheral proinflammatory cytokines such as TNF-α and reactive species such as ROS and RNS are present. Both can cross the BBB to reach the brain. ROS and RNS activate microglia and astrocytes, stimulating them to produce more TNF-α and decreasing AdipoR1 signaling (which is why the levels of AMPK, which is part of this pathway, decrease). TNF-α (which enters the brain from the periphery and is produced by astrocytes and microglia when they are stimulated by ROS and RNS from the periphery), together with the decrease in AMPK, ultimately decrease IRS-1 signaling (IRS-1 is phosphorylated at serine instead of tyrosine, thus decreasing pPI3K and AKT levels). This causes insulin resistance, which is proposed to be the cause of increased Aβ deposition and cognitive loss

Conclusion

The contributions of various transgenic and non-transgenic animal models and inflammatory markers have made it possible to study the relationship between AD and inflammation, a well-known process characteristic of the pathogenesis of AD. Microglia and astrocytes, two main cell types in the CNS, play a critical role in the inflammatory process. These cells release proinflammatory cytokines when they are stimulated by insoluble aggregates such as Aβ and tau. In turn, this response induces synaptic loss and neurodegeneration and further increases Aβ and tau levels, triggering a vicious cycle. Due to their role in inflammation, several conditions, such as obesity, have been implicated as risk factors for AD. Importantly, potential AD treatments that restore the inflammatory phenotype, including the use of NSAIDs, are widely studied and developed. Indeed, clinical, and preclinical trials have already been carried out for this treatment with positive results. Thus, exploring these avenues could modify AD progression and pathology, improving the quality of life of many older adults and their families.

Availability of data and materials

Not applicable.

Abbreviations

AD:

Alzheimer’s disease

AMPK:

AMP-activated protein kinase

Andro:

Andrographolide

APOE:

Apolipoprotein E

APP:

Amyloid precursor protein

BBB:

Blood–brain barrier

BM:

Bone marrow

CNS:

Central nervous system

EVs:

Extracellular vesicles

GFAP:

Glial fibrillar acidic protein

GMF:

Glial maturation factor

GsRb1:

Ginsenoside Rb1

IL-1β:

Interleukin 1β

IL-10:

Interleukin 10

IRS-1:

Insulin receptor substrate 1

LRRs:

Leucine-rich repeats

MHC:

Major histocompatibility complex

NFTs:

Neurofibrillary tangles

NF-κB:

Nuclear factor kappa-activated light chain B

iNOS:

Nitric oxide synthase

NSAIDs:

Nonsteroidal anti-inflammatory drugs

PAR-2:

Protease-activated receptor 2

PBMC:

Peripheral blood mononuclear cell

PDAPP:

Human amyloid precursor protein

PDGF:

Platelet-derived growth factor

PGE2:

Plasma prostaglandin E2

PPARγ:

Peroxisome proliferator-activated receptor γ

ROS:

Reactive oxygen species

RNS:

Reactive nitrogen species

STAT:

Signal transducer and activator of transcription

TNF-α:

Tumor necrosis factor α

TGF-β:

Transforming growth factor beta

TLRs:

Toll-like receptors

WHO:

World Health Organization

References

  1. Pringsheim T, et al. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2014;29(13):1583–90.

    Article  Google Scholar 

  2. Prince M, et al. Recent global trends in the prevalence and incidence of dementia, and survival with dementia. Alzheimer’s Research and Therapy. 2016. https://doi.org/10.1186/s13195-016-0188-8.

    Article  Google Scholar 

  3. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(Suppl 1):S4–9.

    Article  Google Scholar 

  4. Heneka MT, et al. Neuroinflammation in Alzheimer’s disease. The Lancet Neurology. 2015;14(4):388–405.

    Article  CAS  Google Scholar 

  5. Breitner J. Delayed onset of Alzheimer’s disease with nonsteroidal anti-inflammatory and histamine H2 blocking drugs. Neurobiol Aging. 1995;16(4):523–30.

    Article  CAS  Google Scholar 

  6. Barber Pérez P, et al. Evolución del consumo y gasto farmacéutico público de anti-inflamatorios no esteroideos (aines) en el período 2001–2005. Revista Costarricense de Salud Pública. 2007;16(31):19–26.

    Google Scholar 

  7. Lindsay CB, et al. Andrographolide reduces neuroinflammation and oxidative stress in aged Octodon degus. Mol Neurobiol. 2019;57(2):1131–45.

    Article  Google Scholar 

  8. Wang Y, et al. Anti-neuroinflammation effect of ginsenoside Rbl in a rat model of Alzheimer disease. Neurosci Lett. 2011;487(1):70–2.

    Article  CAS  Google Scholar 

  9. Koenigsknecht-Talboo J, et al. Rapid microglial response around amyloid pathology after systemic anti-Abeta antibody administration in PDAPP mice. J Neurosci. 2008;28(52):14156–64.

    Article  CAS  Google Scholar 

  10. Akiyama H, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21(3):383–421.

    Article  CAS  Google Scholar 

  11. Ahmed AU. An overview of inflammation: mechanism and consequences. Front Biol. 2011. https://doi.org/10.1007/s11515-011-1123-9.

    Article  Google Scholar 

  12. Kalden JR. What is inflammation? Eur Heart J. 1987;8(suppl J):1–5.

    Article  Google Scholar 

  13. Chaplin DD. Overview of the immune response. J Allergy Clin Immunol. 2010;125(2 Suppl 2):S3–23.

    Article  Google Scholar 

  14. Fabbri LM, et al. Physiologic consequences of long-term inflammation. Am J Respir Crit Care Med. 1998;157(5):S195–8.

    Article  CAS  Google Scholar 

  15. Libby P. Inflammation and cardiovascular disease mechanisms. Am J Clin Nutr. 2006;83(2):456S-460S.

    Article  CAS  Google Scholar 

  16. Chung HY, et al. Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res Rev. 2009;8(1):18–30.

    Article  CAS  Google Scholar 

  17. Varatharaj A, Galea I. The blood-brain barrier in systemic inflammation. Brain Behav Immun. 2017;60:1–12.

    Article  CAS  Google Scholar 

  18. Carson MJ, Thrash JC, Walter B. The cellular response in neuroinflammation: the role of leukocytes, microglia and astrocytes in neuronal death and survival. Clin Neurosci Res. 2006;6(5):237–45.

    Article  CAS  Google Scholar 

  19. Lull ME, Block ML. Microglial activation and chronic neurodegeneration. Neurotherapeutics. 2010;7(4):354–65.

    Article  CAS  Google Scholar 

  20. Lyman M, et al. Neuroinflammation: the role and consequences. Neurosci Res. 2014;79:1–12.

    Article  CAS  Google Scholar 

  21. Haskó G, et al. Adenosine receptor signaling in the brain immune system. Trends Pharmacol Sci. 2005;26(10):511–6.

    Article  Google Scholar 

  22. Aschner M. Astrocytes as mediators of immune and inflammatory responses in the CNS. Neurotoxicology. 1998;19(2):269–81.

    CAS  Google Scholar 

  23. Loane DJ, Byrnes KR. Role of microglia in neurotrauma. Neurotherapeutics. 2010;7(4):366–77.

    Article  CAS  Google Scholar 

  24. Kaur D, Sharma V, Deshmukh R. Activation of microglia and astrocytes: a roadway to neuroinflammation and Alzheimer’s disease. Inflammopharmacology. 2019;27(4):663–77.

    Article  Google Scholar 

  25. Serrano-Pozo A, et al. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer’s disease. Am J Pathol. 2011;179(3):1373–1373.

    Article  Google Scholar 

  26. Leyns CEG, Holtzman DM. Glial contributions to neurodegeneration in tauopathies. Mol Neurodegener. 2017;12(1):1–16.

    Article  Google Scholar 

  27. Bachiller S, et al. Microglia in neurological diseases: a road map to brain-disease dependent-inflammatory response. Front Cell Neurosci. 2018. https://doi.org/10.3389/fncel.2018.00488.

    Article  Google Scholar 

  28. Nordengen K, et al. Glial activation and inflammation along the Alzheimer’s disease continuum. J Neuroinflammation. 2019. https://doi.org/10.1186/s12974-019-1399-2.

    Article  Google Scholar 

  29. Chun H, et al. Elucidating the interactive roles of glia in Alzheimer’s disease using established and newly developed experimental models. Front Neurol. 2018;9(SEP):797–797.

    Article  Google Scholar 

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

    Article  Google Scholar 

  31. Bolmont T, et al. Dynamics of the microglial/amyloid interaction indicate a role in plaque maintenance. J Neurosci. 2008;28(16):4283–4283.

    Article  CAS  Google Scholar 

  32. Navarro V, et al. Microglia in Alzheimer’s disease: activated, dysfunctional or degenerative. Front Aging Neurosci. 2018. https://doi.org/10.3389/fnagi.2018.00140.

    Article  Google Scholar 

  33. Herber DL, et al. Microglial activation is required for Aβ clearance after intracranial injection of lipopolysaccharide in APP transgenic mice. J Neuroimmune Pharmacol. 2007;2(2):222–31.

    Article  Google Scholar 

  34. Rojo LE, et al. Neuroinflammation: implications for the pathogenesis and molecular diagnosis of Alzheimer’s disease. Arch Med Res. 2008;39(1):1–16.

    Article  CAS  Google Scholar 

  35. Smith JA, et al. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87(1):10–20.

    Article  CAS  Google Scholar 

  36. He P, et al. Deletion of tumor necrosis factor death receptor inhibits amyloid β generation and prevents learning and memory deficits in Alzheimer’s mice. J Cell Biol. 2007;178(5):829–829.

    Article  CAS  Google Scholar 

  37. Siracusa R, Fusco R, Cuzzocrea S. Astrocytes: role and functions in brain pathologies. Front Pharmacol. 2019. https://doi.org/10.3389/fphar.2019.01114.

    Article  Google Scholar 

  38. Pekny M, Pekna M. Astrocyte reactivity and reactive astrogliosis: costs and benefits. Physiol Rev. 2014;94(4):1077–98.

    Article  Google Scholar 

  39. Eng LF, Ghirnikar RS. GFAP and astrogliosis. Brain Pathol. 1994;4(3):229–37.

    Article  CAS  Google Scholar 

  40. Frost GR, Li YM. The role of astrocytes in amyloid production and Alzheimer’s disease. Open Biol. 2017. https://doi.org/10.1098/rsob.170228.

    Article  Google Scholar 

  41. Wyssenbach A, et al. Amyloid β-induced astrogliosis is mediated by β1-integrin via NADPH oxidase 2 in Alzheimer’s disease. Aging Cell. 2016;15(6):1140–52.

    Article  CAS  Google Scholar 

  42. Han X, et al. Astrocyte senescence and Alzheimer’s disease: a review. Front Aging Neurosci. 2020. https://doi.org/10.3389/fnagi.2020.00148.

    Article  Google Scholar 

  43. Verkhratsky A, et al. Astrocytes in Alzheimer’s disease. Neurotherapeutics. 2010;7(4):399–412.

    Article  CAS  Google Scholar 

  44. Pajarillo E, et al. The role of astrocytic glutamate transporters GLT-1 and GLAST in neurological disorders: potential targets for neurotherapeutics. Neuropharmacology. 2019;161:107559–107559.

    Article  CAS  Google Scholar 

  45. Simpson JE, et al. Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain. Neurobiol Aging. 2010;31(4):578–90.

    Article  CAS  Google Scholar 

  46. Kinney JW, et al. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dementia Transl Res Clin Interv. 2018;4:575–575.

    Article  Google Scholar 

  47. Lanfranco GR, et al. Evaluación de la enfermedad de Alzheimer en etapa temprana: biomarcadores y pruebas neuropsicológicas. Rev Med Chil. 2012;140(9):1191–200.

    Article  Google Scholar 

  48. Pascoal TA, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27(9):1592–9.

    Article  CAS  Google Scholar 

  49. Floden AM, Li S, Combs CK. β-Amyloid-stimulated microglia induce neuron death via synergistic stimulation of tumor necrosis factor α and NMDA receptors. J Neurosci. 2005;25(10):2566–2566.

    Article  CAS  Google Scholar 

  50. Streit WJ, Mrak RE, Griffin WST. Microglia and neuroinflammation: a pathological perspective. J Neuroinflammation. 2004;1:14–14.

    Article  Google Scholar 

  51. Rogers J, Lue L-F. Microglial chemotaxis, activation, and phagocytosis of amyloid β-peptide as linked phenomena in Alzheimer’s disease. Neurochem Int. 2001;39(5–6):333–40.

    Article  CAS  Google Scholar 

  52. Liu S, et al. TLR2 is a primary receptor for alzheimer’s amyloid β peptide to trigger neuroinflammatory activation. J Immunol. 2012;188(3):1098–107.

    Article  CAS  Google Scholar 

  53. Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2015;53(2):1181–94.

    Article  Google Scholar 

  54. Fiebich BL, et al. Role of microglia TLRs in neurodegeneration. Front Cell Neurosci. 2018. https://doi.org/10.3389/fncel.2018.00329.

    Article  Google Scholar 

  55. Li C, et al. Inhibitory effects of betulinic acid on LPS-induced neuroinflammation involve M2 microglial polarization via CaMKKβ-dependent AMPK activation. Front Mol Neurosci. 2018;11:98–98.

    Article  Google Scholar 

  56. Chen C, et al. Fasudil regulates T cell responses through polarization of BV-2 cells in mice experimental autoimmune encephalomyelitis. Acta Pharmacol Sin. 2014;35(11):1428–38.

    Article  CAS  Google Scholar 

  57. Escribano L, et al. Rosiglitazone rescues memory impairment in Alzheimer’s transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology. 2010;35(7):1593–604.

    Article  CAS  Google Scholar 

  58. Yamanaka M, et al. PPARγ/RXRα-induced and CD36-mediated microglial amyloid-β phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci. 2012;32(48):17321–31.

    Article  CAS  Google Scholar 

  59. Ji J, et al. Antagonizing peroxisome proliferator-activated receptor γ facilitates M1-to-M2 shift of microglia by enhancing autophagy via the LKB1-AMPK signaling pathway. Aging Cell. 2018;17(4):e12774–e12774.

    Article  Google Scholar 

  60. Yao K, Zu H-B. Microglial polarization: novel therapeutic mechanism against Alzheimer’s disease. Inflammopharmacology. 2019;28(1):95–110.

    Article  Google Scholar 

  61. Ferretti MT, et al. Intracellular Aβ-oligomers and early inflammation in a model of Alzheimer’s disease. Neurobiol Aging. 2012;33(7):1329–42.

    Article  CAS  Google Scholar 

  62. Paranjape GS, et al. Isolated amyloid-β(1–42) protofibrils, but not isolated fibrils, are robust stimulators of microglia. ACS Chem Neurosci. 2012;3(4):302–302.

    Article  CAS  Google Scholar 

  63. Darocha-Souto B, et al. Brain oligomeric β-amyloid but not total amyloid plaque burden correlates with neuronal loss and astrocyte inflammatory response in amyloid precursor protein/tau transgenic mice. J Neuropathol Exp Neurol. 2011;70(5):360–76.

    Article  CAS  Google Scholar 

  64. Crouse NR, et al. Oligomeric amyloid-β(1–42) induces THP-1 human monocyte adhesion and maturation. Brain Res. 2009;1254:109–19.

    Article  CAS  Google Scholar 

  65. Arbel-Ornath M, et al. Soluble oligomeric amyloid-β induces calcium dyshomeostasis that precedes synapse loss in the living mouse brain. Mol Neurodegeneration. 2017. https://doi.org/10.1186/s13024-017-0169-9.

    Article  Google Scholar 

  66. Calvo-Rodriguez M, et al. Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer’s disease. Nat Commun. 2020;11(1):1–17.

    Article  Google Scholar 

  67. Guan PP, et al. Calcium ions aggravate Alzheimer’s disease through the aberrant activation of neuronal networks, leading to synaptic and cognitive deficits. Front Mol Neurosci. 2021. https://doi.org/10.3389/fnmol.2021.757515.

    Article  Google Scholar 

  68. Kierdorf K, Fritz G. RAGE regulation and signaling in inflammation and beyond. J Leukoc Biol. 2013;94(1):55–68.

    Article  CAS  Google Scholar 

  69. Wan W, et al. Aβ1–42 oligomer-induced leakage in an in vitro blood–brain barrier model is associated with up-regulation of RAGE and metalloproteinases, and down-regulation of tight junction scaffold proteins. J Neurochem. 2015;134(2):382–93.

    Article  CAS  Google Scholar 

  70. Sturchler E, et al. Site-specific blockade of RAGE-Vd prevents amyloid-β oligomer neurotoxicity. J Neurosci. 2008;28(20):5149–5149.

    Article  CAS  Google Scholar 

  71. Deane R, et al. A multimodal RAGE-specific inhibitor reduces amyloid β–mediated brain disorder in a mouse model of Alzheimer disease. J Clin Investig. 2012;122(4):1377–92.

    Article  CAS  Google Scholar 

  72. Ferrera D, et al. Resting microglia react to Aβ42 fibrils but do not detect oligomers or oligomer-induced neuronal damage. Neurobiol Aging. 2014;35(11):2444–57.

    Article  CAS  Google Scholar 

  73. Chidambaram H, Das R, Chinnathambi S. Interaction of Tau with the chemokine receptor, CX3CR1 and its effect on microglial activation, migration and proliferation. Cell Biosci. 2020;10:109–109.

    Article  CAS  Google Scholar 

  74. Metcalfe MJ, Figueiredo-Pereira ME. Relationship between tau pathology and neuroinflammation in Alzheimer’s disease. Mount Sinai J Med. 2010;77(1):50–8.

    Article  Google Scholar 

  75. Laurent C, Buée L, Blum D. Tau and neuroinflammation: what impact for Alzheimer’s disease and tauopathies? Biomed J. 2018;41(1):21–33.

    Article  Google Scholar 

  76. Morales G, Farías IG, Maccioni RB. La neuroinflamación como factor detonante del desarrollo de la enfermedad de Alzheimer. Rev Chil Neuropsiquiatr. 2010. https://doi.org/10.4067/S0717-92272010000200007.

    Article  Google Scholar 

  77. Quintanilla RA, et al. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res. 2004. https://doi.org/10.1016/j.yexcr.2004.01.002.

    Article  Google Scholar 

  78. Pawelec P, et al. The impact of the CX3CL1/CX3CR1 axis in neurological disorders. Cells. 2020. https://doi.org/10.3390/cells9102277.

    Article  Google Scholar 

  79. Bolós M, et al. Absence of CX3CR1 impairs the internalization of Tau by microglia. Mol Neurodegener. 2017;12(1):1–14.

    Article  Google Scholar 

  80. Maphis N, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138(6):1738–1738.

    Article  Google Scholar 

  81. Nilson AN, et al. Tau oligomers associate with inflammation in the brain and retina of tauopathy mice and in neurodegenerative diseases. J Alzheimers Dis. 2017;55(3):1083–99.

    Article  CAS  Google Scholar 

  82. Gaikwad S, et al. Tau oligomer induced HMGB1 release contributes to cellular senescence and neuropathology linked to Alzheimer’s disease and frontotemporal dementia. Cell Rep. 2021;36(3):109419–109419.

    Article  CAS  Google Scholar 

  83. Gupta P, et al. Tau oligomers trigger inflammation in the eyes of the Alzhiemer’s disease mouse models. Invest Ophthalmol Vis Sci. 2015;56(7):855–855.

    Google Scholar 

  84. Das R, Balmik AA, Chinnathambi S. Phagocytosis of full-length Tau oligomers by actin-remodeling of activated microglia. J Neuroinflammation. 2020;17(1):1–15.

    Article  Google Scholar 

  85. Sasaki Y, et al. Iba1 is an actin-cross-linking protein in macrophages/microglia. Biochem Biophys Res Commun. 2001;286(2):292–7.

    Article  CAS  Google Scholar 

  86. Deming Y, et al. Triggering receptor expressed on myeloid cells 2 (TREM2): a potential therapeutic target for Alzheimer disease? HHS public access. Expert Opin Ther Targets. 2018;22(7):587–98.

    Article  CAS  Google Scholar 

  87. Atagi Y, et al. Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2). J Biol Chem. 2015;290(43):26043–50.

    Article  CAS  Google Scholar 

  88. Bailey CC, DeVaux LB, Farzan M. The triggering receptor expressed on myeloid cells 2 binds apolipoprotein E. J Biol Chem. 2015;290(43):26033–42.

    Article  CAS  Google Scholar 

  89. Wang Y, et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell. 2015;160(6):1061–71.

    Article  CAS  Google Scholar 

  90. Yeh FL, et al. TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-beta by microglia. Neuron. 2016;91(2):328–40.

    Article  CAS  Google Scholar 

  91. Bekris LM, et al. Soluble TREM2 and biomarkers of central and peripheral inflammation in neurodegenerative disease. J Neuroimmunol. 2018;319:19–27.

    Article  CAS  Google Scholar 

  92. Neumann H, Takahashi K. Essential role of the microglial triggering receptor expressed on myeloid cells-2 (TREM2) for central nervous tissue immune homeostasis. J Neuroimmunol. 2007;184(1–2):92–9.

    Article  CAS  Google Scholar 

  93. Hickman SE, El Khoury J. TREM2 and the neuroimmunology of Alzheimer’s disease. Biochem Pharmacol. 2014;88(4):495–8.

    Article  CAS  Google Scholar 

  94. Doens D, Fernández PL. Microglia receptors and their implications in the response to amyloid β for Alzheimer’s disease pathogenesis. J Neuroinflammation. 2014;11:48–48.

    Article  Google Scholar 

  95. Zhao Y, et al. TREM2 is a receptor for β-amyloid that mediates microglial function. Neuron. 2018;97(5):1023-1031.e7.

    Article  CAS  Google Scholar 

  96. Guerreiro R, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368(2):117–27.

    Article  CAS  Google Scholar 

  97. Jonsson T, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med. 2013;368(2):107–16.

    Article  CAS  Google Scholar 

  98. Zheng H, et al. TREM2 promotes microglial survival by activating Wnt/β-catenin pathway. J Neurosci. 2017;37(7):1772–84.

    Article  CAS  Google Scholar 

  99. Reitz C, Mayeux R. TREM2 and Neurodegenerative Disease. N Engl J Med. 2013;369(16):1564–70.

    Article  CAS  Google Scholar 

  100. Jadhav VS, et al. Trem2 Y38C mutation and loss of Trem2 impairs neuronal synapses in adult mice. Mol Neurodegener. 2020;15(1):62–62.

    Article  CAS  Google Scholar 

  101. Ulrich JD, Holtzman DM. TREM2 function in Alzheimer’s disease and neurodegeneration. ACS Chem Neurosci. 2016;7(4):420–7.

    Article  CAS  Google Scholar 

  102. Wang Y, et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J Exp Med. 2016;213(5):667–75.

    Article  CAS  Google Scholar 

  103. Jay TR, et al. Disease progression-dependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. J Neurosci. 2017;37(3):637–47.

    Article  CAS  Google Scholar 

  104. Jay TR, et al. TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models. J Exp Med. 2015;212(3):287–95.

    Article  CAS  Google Scholar 

  105. Kober DL, et al. Neurodegenerative disease mutations in TREM2 reveal a functional surface and distinct loss-of-function mechanisms. Elife. 2016;5:e20391–e20391.

    Article  Google Scholar 

  106. Bemiller SM, et al. TREM2 deficiency exacerbates tau pathology through dysregulated kinase signaling in a mouse model of tauopathy. Mol Neurodegener. 2017. https://doi.org/10.1186/s13024-017-0216-6.

    Article  Google Scholar 

  107. Jiang T, et al. TREM2 ameliorates neuronal tau pathology through suppression of microglial inflammatory response. Inflammation. 2018;41(3):811–23.

    Article  CAS  Google Scholar 

  108. Leyns CEG, et al. TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci USA. 2017;114(43):11524–9.

    Article  CAS  Google Scholar 

  109. Shi Y, et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature. 2017;549(7673):523–7.

    Article  Google Scholar 

  110. Yuan P, et al. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron. 2016;90(4):724–39.

    Article  CAS  Google Scholar 

  111. Yanamandra K, et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron. 2013;80(2):402–14.

    Article  CAS  Google Scholar 

  112. Komatsu M, et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;441(7095):880–4.

    Article  CAS  Google Scholar 

  113. Gold M, El Khoury J. β-amyloid, microglia, and the inflammasome in Alzheimer’s disease. Semin Immunopathol. 2015;37(6):607–11.

    Article  CAS  Google Scholar 

  114. Ulland TK, et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell. 2017;170(4):649-663.e13.

    Article  CAS  Google Scholar 

  115. Zhao L, Alzheimer A. CD33 in Alzheimer’s disease—biology, pathogenesis, and therapeutics: a mini-review. Gerontology. 2019;65(4):323–31.

    Article  CAS  Google Scholar 

  116. Griciuc A, Tanzi RE. The role of innate immune genes in Alzheimer’s disease. Curr Opin Neurol. 2021;34(2):228–228.

    Article  CAS  Google Scholar 

  117. Griciuc A, et al. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78(4):631–631.

    Article  CAS  Google Scholar 

  118. Magno L, et al. Alzheimer’s disease phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. Alzheimer’s Res Ther. 2019;11(1):1–11.

    Article  Google Scholar 

  119. Tsai AP, et al. PLCG2 is associated with the inflammatory response and is induced by amyloid plaques in Alzheimer’s disease. Genome Medicine. 2022;14(1):1–13.

    Article  Google Scholar 

  120. Jing H, et al. INPP5D rs35349669 polymorphism with late-onset Alzheimer’s disease: a replication study and meta-analysis. Oncotarget. 2016;7(43):69225–69225.

    Article  Google Scholar 

  121. Tsai AP, et al. INPP5D expression is associated with risk for Alzheimer’s disease and induced by plaque-associated microglia. Neurobiol Dis. 2021;153:105303–105303.

    Article  CAS  Google Scholar 

  122. Taipa R, et al. Inflammatory pathology markers (activated microglia and reactive astrocytes) in early and late onset Alzheimer disease: a post mortem study. Neuropathol Appl Neurobiol. 2018;44(3):298–313.

    Article  CAS  Google Scholar 

  123. Ferrari-Souza JP, et al. Astrocyte biomarker signatures of amyloid-β and tau pathologies in Alzheimer’s disease. Mol Psychiatry. 2022;2022:1–9.

    Google Scholar 

  124. Ingelsson M, et al. Early A accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain. Neurology. 2004;62(6):925–31.

    Article  CAS  Google Scholar 

  125. Dinarello CA. Proinflammatory cytokines. Chest. 2000;118(2):503–8.

    Article  CAS  Google Scholar 

  126. Tan ZS, et al. Inflammatory markers and the risk of Alzheimer disease: the Framingham study. Neurology. 2007;68(22):1902–8.

    Article  CAS  Google Scholar 

  127. Chang R, Yee K-L, Sumbria RK. Tumor necrosis factor α inhibition for Alzheimer’s disease. J Cent Nerv Syst Dis. 2017;9:117957351770927–117957351770927.

    Article  Google Scholar 

  128. Kamer AR, et al. TNF-alpha and antibodies to periodontal bacteria discriminate between Alzheimer’s disease patients and normal subjects. J Neuroimmunol. 2009;216(1–2):92–7.

    Article  CAS  Google Scholar 

  129. Tarkowski E, et al. Cerebral pattern of pro- and anti-inflammatory cytokines in dementias. Brain Res Bull. 2003;61(3):255–60.

    Article  CAS  Google Scholar 

  130. Álvarez A, et al. Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in Alzheimer disease. Neurobiol Aging. 2007;28(4):533–6.

    Article  Google Scholar 

  131. Medeiros R, et al. Connecting TNF-alpha signaling pathways to iNOS expression in a mouse model of Alzheimer’s disease: relevance for the behavioral and synaptic deficits induced by amyloid beta protein. J Neurosci. 2007;27(20):5394–404.

    Article  CAS  Google Scholar 

  132. Jones SV, Kounatidis I. Nuclear factor-kappa B and Alzheimer disease, unifying genetic and environmental risk factors from cell to humans. Front Immunol. 2017;8(DEC):1805–1805.

    Article  Google Scholar 

  133. Kaltschmidt B, et al. Transcription factor NF-kappaB is activated in primary neurons by amyloid beta peptides and in neurons surrounding early plaques from patients with Alzheimer disease. Proc Natl Acad Sci USA. 1997;94(6):2642–7.

    Article  CAS  Google Scholar 

  134. Mattson MP, Camandola S. NF-κB in neuronal plasticity and neurodegenerative disorders. J Clin Investig. 2001;107(3):247–247.

    Article  CAS  Google Scholar 

  135. Barger SW, Harmon AD. Microglial activation by Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature. 1997;388(6645):878–81.

    Article  CAS  Google Scholar 

  136. Chen C-H, et al. Increased NF-κB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int J Neuropsychopharmacol. 2011;15(01):77–90.

    Article  Google Scholar 

  137. Decourt B, Lahiri DK, Sabbagh MN. Targeting tumor necrosis factor alpha for Alzheimer’s disease. Curr Alzheimer Res. 2017;14(4):412–412.

    Article  CAS  Google Scholar 

  138. Wang C, et al. Microglial NF-κB drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun. 2022. https://doi.org/10.1038/s41467-022-29552-6.

    Article  Google Scholar 

  139. Crews L, Masliah E. Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum Mol Genet. 2010;19(R1):R12–20.

    Article  CAS  Google Scholar 

  140. Selles MC, Oliveira MM, Ferreira ST. Brain inflammation connects cognitive and non-cognitive symptoms in Alzheimer’s disease. J Alzheimers Dis. 2018;64(s1):S313–27.

    Article  Google Scholar 

  141. Chen W-W, Zhang X, Huang W-J. Role of neuroinflammation in neurodegenerative diseases (review). Mol Med Rep. 2016;13(4):3391–6.

    Article  CAS  Google Scholar 

  142. Young JK. Neurogenesis makes a crucial contribution to the neuropathology of Alzheimer’s disease. J Alzheimer’s Dis Rep. 2020;4(1):365–365.

    Article  Google Scholar 

  143. Kempuraj D, et al. Neuroinflammation induces neurodegeneration. J Neurol Neurosurg Spine. 2016;1(1):1003.

    Google Scholar 

  144. Huang X, Hussain B, Chang J. Peripheral inflammation and blood–brain barrier disruption: effects and mechanisms. CNS Neurosci Ther. 2021;27(1):36–36.

    Article  CAS  Google Scholar 

  145. Coelho AA, et al. Inducible nitric oxide synthase inhibition in the medial prefrontal cortex attenuates the anxiogenic-like effect of acute restraint stress via CB1 receptors. Front Psych. 2022;13:1359–1359.

    Google Scholar 

  146. Di Filippo M, et al. Mitochondria and the link between neuroinflammation and neurodegeneration. J Alzheimers Dis. 2010;20(s2):S369–79.

    Article  Google Scholar 

  147. Calkins MJ, Manczak M, Reddy PH. Mitochondria-targeted antioxidant SS31 prevents amyloid beta-induced mitochondrial abnormalities and synaptic degeneration in Alzheimer’s disease. Pharmaceuticals. 2012;5(10):1103–1103.

    Article  CAS  Google Scholar 

  148. Picone P, et al. Mitochondrial dysfunction: different routes to Alzheimer’s disease therapy. Oxid Med Cell Longev. 2014;2014:780179–780179.

    Article  Google Scholar 

  149. Wang B, et al. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther. 2021;6(1):1–30.

    Google Scholar 

  150. Rao JS, et al. Neuroinflammation and synaptic loss. Neurochem Res. 2012;37(5):903–10.

    Article  CAS  Google Scholar 

  151. Julien C, et al. Decreased drebrin mRNA expression in Alzheimer disease: correlation with tau pathology. J Neurosci Res. 2008;86(10):2292–302.

    Article  CAS  Google Scholar 

  152. Popp J, et al. Macrophage migration inhibitory factor in mild cognitive impairment and Alzheimer’s disease. J Psychiatr Res. 2009;43(8):749–53.

    Article  Google Scholar 

  153. Rao C, Reddy B. NSAIDs and chemoprevention. Curr Cancer Drug Targets. 2004;4(1):29–42.

    Article  CAS  Google Scholar 

  154. O’Bryant SE, et al. A precision medicine model for targeted NSAID therapy in Alzheimer’s disease. J Alzheimers Dis. 2018;66(1):97–97.

    Article  CAS  Google Scholar 

  155. Breitner JCS, et al. Risk of dementia and AD with prior exposure to NSAIDs in an elderly community-based cohort. Neurology. 2009;72(22):1899–905.

    Article  CAS  Google Scholar 

  156. McGeer PL, McGeer EG. NSAIDs and Alzheimer disease: epidemiological, animal model and clinical studies. Neurobiol Aging. 2007;28(5):639–47.

    Article  CAS  Google Scholar 

  157. Zandi P. Do NSAIDs prevent Alzheimer’s disease? And, if so, why? The epidemiological evidence. Neurobiol Aging. 2001;22(6):811–7.

    Article  CAS  Google Scholar 

  158. Heneka MT, et al. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain. 2005;128(Pt 6):1442–53.

    Article  Google Scholar 

  159. Choi SH, et al. Cyclooxygenase-1 inhibition reduces amyloid pathology and improves memory deficits in a mouse model of Alzheimer’s disease. J Neurochem. 2013;124(1):59–59.

    Article  CAS  Google Scholar 

  160. Moore AH, et al. Non-steroidal anti-inflammatory drugs in alzheimer’s disease and Parkinson’s disease: reconsidering the role of neuroinflammation. Pharmaceuticals. 2010;3(6):1812–41.

    Article  CAS  Google Scholar 

  161. Imbimbo BP, Solfrizzi V, Panza F. Are NSAIDs useful to treat Alzheimer’s disease or mild cognitive impairment? Front Aging Neurosci. 2010;2:19–19.

    CAS  Google Scholar 

  162. Eriksen JL, et al. NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Investig. 2003;112(3):440–9.

    Article  CAS  Google Scholar 

  163. Hershey LA, Lipton RB. Naproxen for presymptomatic Alzheimer disease. Neurology. 2019;92(18):829–30.

    Article  Google Scholar 

  164. Martin B. Double placebo design in a prevention trial for Alzheimer’s disease. Control Clin Trials. 2002;23(1):93–9.

    Article  Google Scholar 

  165. Barranco Quintana JL, et al. Factores de riesgo de la enfermedad de Alzheimer. Revista de Neurología. 2005;40(10):613–613.

    Article  CAS  Google Scholar 

  166. Guan PP, et al. Cyclooxygenase-2 induced the β-amyloid protein deposition and neuronal apoptosis via upregulating the synthesis of prostaglandin E2 and 15-deoxy-Δ12,14-prostaglandin J2. Neurotherapeutics. 2019;16(4):1255–1255.

    Article  CAS  Google Scholar 

  167. Yermakova AV, et al. Cyclooxygenase-1 in human alzheimer and control brain: quantitative analysis of expression by microglia and CA3 hippocampal neurons. J Neuropathol Exp Neurol. 1999;58(11):1135–46.

    Article  CAS  Google Scholar 

  168. Schatzberg Morón AF, Nemeroff CB. Tratado de psicofarmacología. Barcelona: Editorial Médica Panamericana SA; 2006.

    Google Scholar 

  169. McAdam BF, et al. Systemic biosynthesis of prostacyclin by cyclooxygenase (COX)-2: the human pharmacology of a selective inhibitor of COX-2. Proc Natl Acad Sci. 1999;96(1):272–7.

    Article  CAS  Google Scholar 

  170. Prasad KN, et al. Prostaglandins as putative neurotoxins in Alzheimer’s disease. Exp Biol Med. 2016;219(2):120–5. https://doi.org/10.3181/00379727-219-44323.

    Article  Google Scholar 

  171. Elder GA, Gama Sosa MA, De Gasperi R. Transgenic mouse models of Alzheimer’s disease. Mount Sinai J Med. 2010;77(1):69–69.

    Article  Google Scholar 

  172. Chen G, et al. A learning deficit related to age and β-amyloid plaques in a mouse model of Alzheimer’s disease. Nature. 2000;408(6815):975–9.

    Article  CAS  Google Scholar 

  173. Götz J, et al. Transgenic animal models of Alzheimer’s disease and related disorders: histopathology, behavior and therapy. Mol Psychiatry. 2004;9(7):664–83.

    Article  Google Scholar 

  174. Deters N, Ittner LM, Götz J. Divergent phosphorylation pattern of tau in P301L tau transgenic mice. Eur J Neurosci. 2008;28(1):137–47.

    Article  Google Scholar 

  175. Lewis J, et al. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet. 2000;25(4):402–5.

    Article  CAS  Google Scholar 

  176. Cisternas P, et al. New insights into the spontaneous human Alzheimer’s disease-like model Octodon degus: unraveling amyloid-β peptide aggregation and age-related amyloid pathology. J Alzheimers Dis. 2018;66(3):1145–63.

    Article  CAS  Google Scholar 

  177. Inestrosa NC, et al. Human-like rodent amyloid-β-peptide determines Alzheimer pathology in aged wild-type Octodon degu. Neurobiol Aging. 2005;26(7):1023–8.

    Article  CAS  Google Scholar 

  178. Shankar GM, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42.

    Article  CAS  Google Scholar 

  179. Wilcox KC, et al. Aβ oligomer-induced synapse degeneration in Alzheimer’s disease. Cell Mol Neurobiol. 2011;31(6):939–48.

    Article  CAS  Google Scholar 

  180. Sarasa M, Pesini P. Natural non-trasgenic animal models for research in alzheimer’s disease. Curr Alzheimer Res. 2009;6(2):171–78. https://doi.org/10.2174/156720509787602834

    Article  CAS  Google Scholar 

  181. Kim JH, et al. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res. 2017;41(4):435–43.

    Article  Google Scholar 

  182. Brown ML, Schneyer A. A decade later: revisiting the TGFβ family’s role in diabetes. Trends Endocrinol Metab. 2021;32(1):36–47.

    Article  CAS  Google Scholar 

  183. Lye TC, Shores EA. Traumatic brain injury as a risk factor for Alzheimer’s disease: a review. Neuropsychol Rev. 2000;10(2):115–29.

    Article  CAS  Google Scholar 

  184. Ríos JA, et al. Is Alzheimer’s disease related to metabolic syndrome? A Wnt signaling conundrum. Prog Neurobiol. 2014;121:125–46.

    Article  Google Scholar 

  185. Sastre M, et al. Inflammatory risk factors and pathologies associated with Alzheimers disease. Curr Alzheimer Res. 2011;8(2):132–41.

    Article  CAS  Google Scholar 

  186. Sims-Robinson C, et al. How does diabetes accelerate Alzheimer disease pathology? Nat Rev Neurol. 2010;6(10):551–9.

    Article  CAS  Google Scholar 

  187. Zhang Q, et al. Risk prediction of late-onset Alzheimer’s disease implies an oligogenic architecture. Nat Commun. 2020;11(1):4799–4799.

    Article  CAS  Google Scholar 

  188. Bracko O, et al. High fat diet worsens Alzheimer’s disease-related behavioral abnormalities and neuropathology in APP/PS1 mice, but not by synergistically decreasing cerebral blood flow. Sci Rep. 2020;10(1):1–16.

    Article  Google Scholar 

  189. NCD Risk Factor Collaboration (NCD-RisC). Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19·2 million participants. Lancet. 2016;387(10026):1377–96. https://doi.org/10.1016/S0140-6736(16)30054-X.

    Article  Google Scholar 

  190. Park HS, Park JY, Yu R. Relationship of obesity and visceral adiposity with serum concentrations of CRP, TNF-α and IL-6. Diabetes Res Clin Pract. 2005;69(1):29–35.

    Article  CAS  Google Scholar 

  191. Hahm JR, et al. Metabolic stress alters antioxidant systems, suppresses the adiponectin receptor 1 and induces Alzheimer’s like pathology in mice brain. Cells. 2020;9(1):249–249.

    Article  CAS  Google Scholar 

  192. Caterson ID, Gill TP. Obesity: epidemiology and possible prevention. Best Pract Res Clin Endocrinol Metab. 2002;16(4):595–610.

    Article  Google Scholar 

  193. Cisternas P, et al. Modulation of glucose metabolism in hippocampal neurons by adiponectin and resistin. Mol Neurobiol. 2019;56(4):3024–37.

    Article  CAS  Google Scholar 

  194. Aragonès G, et al. Circulating microbiota-derived metabolites: a "liquid biopsy? Int J Obes. 2020;44(4):875–85.

    Article  Google Scholar 

  195. Heger LA, et al. Inflammation in acute coronary syndrome: expression of TLR2 mRNA is increased in platelets of patients with ACS. PLoS ONE. 2019;14(10):e0224181–e0224181.

    Article  CAS  Google Scholar 

  196. McDonald CL, et al. Inhibiting TLR2 activation attenuates amyloid accumulation and glial activation in a mouse model of Alzheimer’s disease. Brain Behav Immun. 2016;58:191–200.

    Article  CAS  Google Scholar 

  197. Pourbadie HG, et al. Early minor stimulation of microglial TLR2 and TLR4 receptors attenuates Alzheimer’s disease-related cognitive deficit in rats: behavioral, molecular, and electrophysiological evidence. Neurobiol Aging. 2018;70:203–16.

    Article  CAS  Google Scholar 

  198. Re F, Strominger JL. Toll-like receptor 2 (TLR2) and TLR4 differentially activate human dendritic cells. J Biol Chem. 2001;276(40):37692–9.

    Article  CAS  Google Scholar 

  199. Szymańska A, et al. TLR2 expression on leukemic B cells from patients with chronic lymphocytic leukemia. Arch Immunol Ther Exp. 2019;67(1):55–65.

    Article  Google Scholar 

  200. Butovsky O, et al. Activation of microglia by aggregated β-amyloid or lipopolysaccharide impairs MHC-II expression and renders them cytotoxic whereas IFN-γ and IL-4 render them protective. Mol Cell Neurosci. 2005;29(3):381–93.

    Article  CAS  Google Scholar 

  201. Lin J, et al. PD-1+CXCR5−CD4+T cells are correlated with the severity of systemic lupus erythematosus. Rheumatology. 2019;58(12):2188–92.

    Article  CAS  Google Scholar 

  202. Mittal K, et al. CD4 T cells induce a subset of MHCII-expressing microglia that attenuates Alzheimer pathology. iScience. 2019;16:298–311.

    Article  CAS  Google Scholar 

  203. Pabon MM, et al. CX3CL1 reduces neurotoxicity and microglial activation in a rat model of Parkinson’s disease. J Neuroinflammation. 2011;8:9–9.

    Article  CAS  Google Scholar 

  204. Smith HJ, et al. The antitumor effects of entinostat in ovarian cancer require adaptive immunity. Cancer. 2018;124(24):4657–66.

    Article  CAS  Google Scholar 

  205. Trier N, et al. Human MHC-II with shared epitope motifs are optimal Epstein-Barr virus glycoprotein 42 ligands-relation to rheumatoid arthritis. Int J Mol Sci. 2018;19(1):317–317.

    Article  Google Scholar 

  206. Ahmed Z, et al. Actin-binding proteins coronin-1a and IBA-1 are effective microglial markers for immunohistochemistry. J Histochem Cytochem. 2007;55(7):687–700.

    Article  CAS  Google Scholar 

  207. de Zorzi VN, et al. Galangin prevents increased susceptibility to pentylenetetrazol-stimulated seizures by prostaglandin E2. Neuroscience. 2019;413:154–68.

    Article  Google Scholar 

  208. Sillerud LO, et al. Longitudinal monitoring of microglial/macrophage activation in ischemic rat brain using Iba-1-specific nanoparticle-enhanced magnetic resonance imaging. J Cereb Blood Flow Metab. 2020;40(1_suppl):S117–33.

    Article  CAS  Google Scholar 

  209. Neri M, et al. Immunohistochemical evaluation of aquaporin-4 and its correlation with CD68, IBA-1, HIF-1α, GFAP, and CD15 expressions in fatal traumatic brain injury. Int J Mol Sci. 2018;19(11):3544–3544.

    Article  Google Scholar 

  210. Qiu L-L, et al. Dysregulation of BDNF/TrkB signaling mediated by NMDAR/Ca(2+)/calpain might contribute to postoperative cognitive dysfunction in aging mice. J Neuroinflammation. 2020;17(1):23–23.

    Article  Google Scholar 

  211. Sakae N, et al. Microglia in frontotemporal lobar degeneration with progranulin or C9ORF72 mutations. Ann Clin Transl Neurol. 2019;6(9):1782–96.

    Article  CAS  Google Scholar 

  212. Salobrar-García E, et al. Microglial activation in the retina of a triple-transgenic Alzheimer’s disease mouse model (3xTg-AD). Int J Mol Sci. 2020;21(3):816–816.

    Article  Google Scholar 

  213. Abdelhak A, et al. Serum GFAP as a biomarker for disease severity in multiple sclerosis. Sci Rep. 2018;8(1):14798–14798.

    Article  CAS  Google Scholar 

  214. Eng LF, Ghirnikar RS, Lee YL. Glial fibrillary acidic protein: GFAP-thirty-one years (1969–2000). Neurochem Res. 2000;25(9):1439–51.

    Article  CAS  Google Scholar 

  215. Kamphuis W, et al. GFAP isoforms in adult mouse brain with a focus on neurogenic astrocytes and reactive astrogliosis in mouse models of Alzheimer disease. PLoS ONE. 2012;7(8):e42823–e42823.

    Article  CAS  Google Scholar 

  216. Hagemann TL, et al. Antisense suppression of glial fibrillary acidic protein as a treatment for Alexander disease. Ann Neurol. 2018;83(1):27–39.

    Article  CAS  Google Scholar 

  217. Iorio R, et al. Clinical and immunological characteristics of the spectrum of GFAP autoimmunity: a case series of 22 patients. J Neurol Neurosurg Psychiatry. 2017;89(2):138–46.

    Article  Google Scholar 

  218. Kimura A, et al. Clinical characteristics of autoimmune GFAP astrocytopathy. J Neuroimmunol. 2019;332:91–8.

    Article  CAS  Google Scholar 

  219. Nichols NR, et al. GFAP mRNA increases with age in rat and human brain. Neurobiol Aging. 1993;14(5):421–9.

    Article  CAS  Google Scholar 

  220. Yue JK, et al. Association between plasma GFAP concentrations and MRI abnormalities in patients with CT-negative traumatic brain injury in the TRACK-TBI cohort: a prospective multicentre study. The Lancet Neurology. 2019;18(10):953–61.

    Article  CAS  Google Scholar 

  221. Chen Y-L, et al. Serum TNF-α concentrations in type 2 diabetes mellitus patients and diabetic nephropathy patients: a systematic review and meta-analysis. Immunol Lett. 2017;186:52–8.

    Article  CAS  Google Scholar 

  222. Corrado A, et al. Anti-TNF-α effects on anemia in rheumatoid and psoriatic arthritis. Int J Immunopathol Pharmacol. 2017;30(3):302–7.

    Article  CAS  Google Scholar 

  223. Furue K, et al. Psoriasis and the TNF/IL23/IL17 axis. G Ital Dermatol Venereol. 2019. https://doi.org/10.23736/S0392-0488.18.06202-8.

    Article  Google Scholar 

  224. Starkie R, et al. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-α production in humans. FASEB J. 2003;17(8):1–10.

    Article  Google Scholar 

  225. Zhang Y-Y, et al. Atorvastatin attenuates the production of IL-1β, IL-6, and TNF-α in the hippocampus of an amyloid β1-42-induced rat model of Alzheimer’s disease. Clin Interv Aging. 2013;8:103–10.

    CAS  Google Scholar 

  226. Afsar B, et al. The future of IL-1 targeting in kidney disease. Drugs. 2018;78(11):1073–83.

    Article  CAS  Google Scholar 

  227. Buckley LF, Abbate A. Interleukin-1 blockade in cardiovascular diseases: a clinical update. Eur Heart J. 2018;39(22):2063–9.

    Article  CAS  Google Scholar 

  228. Colton CA, et al. Expression profiles for macrophage alternative activation genes in AD and in mouse models of AD. J Neuroinflammation. 2006;3:27–27.

    Article  Google Scholar 

  229. Herder C, et al. The IL-1 pathway in type 2 diabetes and cardiovascular complications. Trends Endocrinol Metab. 2015;26(10):551–63.

    Article  CAS  Google Scholar 

  230. Toplak N, Blazina Š, Avčin T. The role of IL-1 inhibition in systemic juvenile idiopathic arthritis: current status and future perspectives. Drug Des Dev Ther. 2018;12:1633–43.

    Article  CAS  Google Scholar 

  231. Ahmed AS, et al. NF-κB-associated pain-related neuropeptide expression in patients with degenerative disc disease. Int J Mol Sci. 2019;20(3):658–658.

    Article  CAS  Google Scholar 

  232. Gao J, et al. Salidroside suppresses inflammation in a D-galactose-induced rat model of Alzheimer’s disease via SIRT1/NF-κB pathway. Metab Brain Dis. 2016;31(4):771–8.

    Article  CAS  Google Scholar 

  233. Lukiw WJ, Zhao Y, Cui JG. An NF-kappaB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem. 2008;283(46):31315–22.

    Article  CAS  Google Scholar 

  234. Plantinga TS, et al. Association of NF-κB polymorphisms with clinical outcome of non-medullary thyroid carcinoma. Endocr Relat Cancer. 2017. https://doi.org/10.1530/ERC-17-0033.

    Article  Google Scholar 

  235. Soleimani A, et al. Role of the NF-κB signaling pathway in the pathogenesis of colorectal cancer. Gene. 2020;726:144132–144132.

    Article  CAS  Google Scholar 

  236. Zhang F, et al. Acute hypoxia induced an imbalanced M1/M2 activation of microglia through NF-κB signaling in Alzheimer’s disease mice and wild-type littermates. Front Aging Neurosci. 2017;9:282–282.

    Article  Google Scholar 

  237. Zheng L, et al. Placental expression of AChE, α7nAChR and NF-κB in patients with preeclampsia. Ginekol Pol. 2018;89(5):249–55.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Basal Center of Excellence in Aging and Regeneration (ANID-ACE210009) to NCI. We also thank the Sociedad Química y Minera de Chile (SQM) for the special Grant “The Role of Lithium in Human Health and Disease” and the European Union and FONIS-ANID-Chile for the Grants “High fat diet, microbiota and neuroinflammation in the progression of Alzheimer disease” (EULACH16/T010131) and “Mechanistic insights into Alzheimer’s disease: epigenetic and immunomodulatory remodeling of endocannabinoid signaling” (EULACH16/T010132), all of which were given to NCI.

Author information

Authors and Affiliations

Authors

Contributions

NCI and CN contributed to the conception of the review; this work was conceived during the Research Unit of the BIO-296 Undergraduate Course of CN. CN, PS, PC, CG, RVS, JMZ and NCI contributed to the writing of the manuscript. PS, CG and NCI edited the final draft of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Nibaldo C. Inestrosa.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The authors approved this version of the manuscript to publish.

Competing interests

The authors have nothing to disclose.

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

Novoa, C., Salazar, P., Cisternas, P. et al. Inflammation context in Alzheimer’s disease, a relationship intricate to define. Biol Res 55, 39 (2022). https://doi.org/10.1186/s40659-022-00404-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40659-022-00404-3