Skip to main content

A small-molecule TLR4 antagonist reduced neuroinflammation in female E4FAD mice

Abstract

Background

APOE genotype is the greatest genetic risk factor for sporadic Alzheimer’s disease (AD). APOE4 increases AD risk up to 12-fold compared to APOE3, an effect that is greater in females. Evidence suggests that one-way APOE could modulate AD risk and progression through neuroinflammation. Indeed, APOE4 is associated with higher glial activation and cytokine levels in AD patients and mice. Therefore, identifying pathways that contribute to APOE4-associated neuroinflammation is an important approach for understanding and treating AD. Human and in vivo evidence suggests that TLR4, one of the key receptors involved in the innate immune system, could be involved in APOE-modulated neuroinflammation. Consistent with that idea, we previously demonstrated that the TLR4 antagonist IAXO-101 can reduce LPS- and Aβ-induced cytokine secretion in APOE4 glial cultures. Therefore, the goal of this study was to advance these findings and determine whether IAXO-101 can modulate neuroinflammation, Aβ pathology, and behavior in mice that express APOE4.

Methods

We used mice that express five familial AD mutations and human APOE3 (E3FAD) or APOE4 (E4FAD). Female and male E4FAD mice and female E3FAD mice were treated with vehicle or IAXO-101 in two treatment paradigms: prevention from 4 to 6 months of age or reversal from 6 to 7 months of age. Learning and memory were assessed by modified Morris water maze. Aβ deposition, fibrillar amyloid deposition, astrogliosis, and microgliosis were assessed by immunohistochemistry. Soluble levels of Aβ and apoE, insoluble levels of apoE and Aβ, and IL-1β were measured by ELISA.

Results

IAXO-101 treatment resulted in lower Iba-1 coverage, lower number of reactive microglia, and improved memory in female E4FAD mice in both prevention and reversal paradigms. IAXO-101-treated male E4FAD mice also had lower Iba-1 coverage and reactivity in the RVS paradigm, but there was no effect on behavior. There was also no effect of IAXO-101 treatment on neuroinflammation and behavior in female E3FAD mice.

Conclusion

Our data supports that TLR4 is a potential mechanistic therapeutic target for modulating neuroinflammation and cognition in APOE4 females.

Background

APOE (apolipoprotein E) genotype is a major risk factor for sporadic Alzheimer’s disease (AD), with APOE4 increasing risk up to 12-fold compared to APOE3 (reviewed in [1, 2]). Therefore, an important challenge is identifying therapeutic targets for APOE4 carriers in AD. The role of APOE in AD is complex, as APOE has been shown to modulate multiple functions and pathways in humans and transgenic mouse models that overproduce amyloid-beta (Aβ) via familial AD mutations (FAD) (reviewed in [3, 4]). For example, compared to APOE3, APOE4 is associated with higher soluble Aβ levels and amyloid plaques, altered metabolism, and neurovascular function (reviewed in [3,4,5]). In addition, increasing evidence suggests that APOE-modulated neuroinflammation contributes to AD progression [6]. In AD patients, compared to APOE3, APOE4 is associated with earlier onset of cognitive deficits and increased neuroinflammation [7,8,9,10]. These human data are recapitulated in vivo, as APOE4 is associated with greater microgliosis [11,12,13,14], astrogliosis [13,14,15], and altered cytokine levels [12, 14, 16, 17] in FAD mice and in APOE-knock in mice after induction of peripheral inflammation. In addition, there is greater neuroinflammation in female APOE4 FAD mice compared to males [11, 15, 18, 19], consistent with higher AD risk and pathology in female APOE4 carriers. Therefore, identifying pathways that contribute to APOE4-associated neuroinflammation is an important approach for developing AD therapeutics.

Neuroinflammation is complex and involves multiple cell-types, receptors, signaling pathways, cytokines, and chemokines. One approach to identify the contribution of neuroinflammatory pathways to AD progression is to evaluate activity of FDA approved drugs with known anti-inflammatory activities such as NSAIDs [20]. Although some studies supported that NSAIDs may be efficacious as an AD therapeutic, including in APOE4 carriers, others have shown no beneficial effects [21,22,23]. Ongoing research is defining if these established anti-inflammatories will be beneficial for specific patient groups at certain stages of AD and the optimal treatment regime. An alternative approach is to determine if compounds that target pathways modulated by APOE4 can impact neuroinflammation and behavior. The innate immune system is one of the most highly conserved immune responses across plants, drosophila, and animals to defend against invading pathogens and is also activated by endogenous stress-related molecules. Toll-like receptor 4 (TLR4) is a key component of the innate immune response is expressed by microglia (and to a lesser extent by astrocytes) [24, 25] and is activated by lipopolysaccharide (LPS) and other stress-associated ligands (reviewed in [26, 27]). Evidence suggests that TLR4 could contribute to neuroinflammation in AD. For example, there is higher TLR4 expression in AD patients’ [28, 29] and FAD mouse brains [29, 30] and single nucleotide polymorphisms (SNPs) in TLR4 have been shown to modulate AD risk [31,32,33]. There is also evidence that TLR4 is involved in APOE4-associated neuroinflammation. In vitro, LPS-induced inflammatory response is greater with APOE4 compared to APOE3 [14, 16, 17]. Furthermore, Aβ-induced cytokine production is greater with APOE4 in glial cultures, an effect that is blocked by TLR4 antagonists, and expression of TLR4-related genes are greater in APOE4-FAD mice [34]. Based on these data, it has been proposed that with APOE4, greater TLR4 activation in the brain may lead to higher neuroinflammatory responses and contribute to behavioral deficits. However, to date, no studies have directly evaluated the effect of blocking TLR4 on AD-relevant pathology and behavior in mice that express APOE4.

The goal of this study was to determine whether TLR4 antagonism can modulate neuroinflammation, Aβ pathology, and behavior in mice that express APOE4. To address this goal, we used a novel small molecule TLR4 antagonist IAX0-101, which we have previously shown to inhibit Aβ-induced cytokine release in APOE4-mixed glial cells [34]. Therefore, we treated mice that express APOE4 (E4FAD) and overproduce Aβ with IAXO-101 in prevention and reversal paradigms and evaluated the impact on neuroinflammation, Aβ pathology, and behavior.

Methods

Animals and study design

All experiments follow the University of Illinois at Chicago Animal Care Committee protocols. EFAD mice express five familial AD mutations and human APOE (5xFAD+/-/hAPOE+/+) as described in [35]. EFAD mice were generated from Tg6799, the 5xFAD mouse strain that produced the highest amount of Aβ42. In Tg6799 5xFAD mice, Aβ40 levels also increase with age but rise more slowly and are substantially lower than for Aβ42 in young mice [36]. The prevention (PVT) treatment paradigm was designed to begin at 4 months, at early stages of AD pathology, including amyloid deposition and neuroinflammation, and end at 6 months when pathology is significant [35]. The reversal (RVS) treatment paradigm begins at 6 months and ends at 7 months, capturing the previously observed age-associated increase in pathology [37].

Drug formulation and treatment

IAXO-101, a synthetic Cluster of differentiation 14 (CD14)/TLR4 antagonist nano-formulated in Lipodisq™, was provided by Innaxon Biosciences (Tewkesbury, UK). Lipodisq™ drug formulations are lipid-based nano-sized (10-40 nm) monodisperse, discoidal nanoparticles, also referred to as native nano-discs or styrene maleic acid lipid particles [38]. EFAD mice were administered either IAXO-101 in Lipodisq™ nano-formulation or a vehicle (empty Lipodisq™ nano-formulation) using subcutaneous injections at 10 mg/kg for three times per week. The nano-formulated IAXO-101 was provided as a sterile, endotoxin-tested, 4 mg/ml aqueous stock solution, diluted just prior to use in sterile, endotoxin-free water [38, 39]. Treatments for the mice were randomized within cage and across groups. All investigators were blinded for treatment and analysis. Body weights were measured prior to each injection to determine dose and monitor for any treatment-related weight changes.

Behavioral analysis and tissue harvest

All behavioral data were recorded and analyzed with ANY-maze video tracking software (Stoelting Co, Wood Dale, IL USA). In the week prior to sacrifice, mouse behavior was tested using a modified Morris water maze protocol with acquisition trials consisting of 4 × 1 min trials/day for 5 consecutive days with latency to the platform recorded for each trial. A single probe trial was run on day 6 with the platform removed, and the readouts included latency to platform and latency to target quadrant (previously described [40,41,42]). After the probe trial, the mice were anesthetized with ketamine/xylazine and perfused with phosphate-buffered saline. Then, the brains were removed and dissected at the midline to produce two hemi-brains, one each for immunohistochemical and biochemical analysis (previously described in [35, 42]).

Immunohistochemical analysis

Serial sagittal brain sections (35 μm thick, 280 μm apart) from EFAD mice were all stained for fibrillar amyloid deposition via Thio-S and immunostained for Aβ deposition, astrogliosis, and microgliosis (previously described [11, 14, 35]), with anti-mouse or anti-rabbit Alexa-fluor secondary antibodies. A list of all the antibodies used in this study is provided in Additional file 2. The stained sections were imaged at × 10 magnification with a Zeiss Fluorescence microscope and analyzed for area covered by Thio-S, MOAB-2, GFAP, S100β, C3, Iba-1, and Clec7a in the cortex (CX) and hippocampus (HP) using ImageJ. The regions included for analysis are outlined in Fig. S1A (CX = 1; HP = 2 + 3) and close-up images of CX and HP at × 20 for all immunostainings performed are presented in Additional file 1 (Figs. S8-S15). Immunostaining signals from both cortical and hippocampal regions were quantified by investigators blinded to treatment, APOE genotype, and sex within paradigm. For morphological analysis of microglia, 8 sub-regions within the CX were imaged at × 40, and the total number of microglia within each frame was classified as type 1 or type 2/3 (Fig. S1B) [43].

Sequential protein extraction fractions and ELISAs (Aβ, apoE, and IL-1β)

Frozen cortices dissected from the mouse hemi-brains were homogenized using a three-step extraction protocol producing soluble (Tris-buffered saline: TBS), non-ionic detergent (TBS + 1%Triton X-100: TBSX), and insoluble (neutralized formic acid: FA) [44]. Total protein in the TBS and insoluble extracts was quantified using the Bradford assay [44]. Soluble Aβ42, apoE, and interleukin-1 beta (IL-1β) were measured by ELISA following the manufacturer’s instructions, while insoluble Aβ42 and apoE were measured in insoluble fraction (previously described [12, 35, 44, 45]). A list of all the antibodies used is provided in Additional file 2.

Statistical analysis

GraphPad Prism 9 (for Mac, GraphPad Software, La Jolla, CA) was used for statistical analyses. For all statistical tests, p < 0.05 was considered significant. Data was plotted as scatter bar graphs, with the mean and standard error of the mean (SEM). Morris water maze acquisition phase data and body weights were analyzed by repeated measured two- or three-way ANOVA, followed by Tukey’s post hoc tests. All other data were analyzed by unpaired Student’s t-test. See Additional file 3 for details on n sizes and statistical comparisons.

Results

The goal of this study was to evaluate the effect of TLR4 antagonism on neuroinflammation and other markers of AD-relevant pathology in mice that express human APOE. To address this goal, we treated EFAD mice from 4 to 6 months of age (Prevention paradigm; PVT) or from 6 to 7 months of age (reversal paradigm; RVS) with either vehicle or IAXO-101 (10 mg/kg ~ 0.3 mg/mouse, 3 subcutaneous injections/week). We used EFAD mice as they overproduce Aβ42 and express human APOE4 (E4FAD) or APOE3. Previous data have demonstrated that E4FAD mice have higher levels of astrogliosis, microgliosis, and Aβ pathology at 6 months of age compared to E3FAD mice [12, 35]. Therefore, in our prevention paradigm, we treated mice from 4 to 6 months or age, and in our reversal paradigm, treatment was from 6 to 7 months. For TLR4 antagonism, we used IAXO-101, as we have previously demonstrated that this compound lowers inflammation in vitro using APOE4 mixed glial cultures. IAXO-101 has been used in vivo at doses ranging from 0.06 to 0.3 mg/mouse/day via various routes [46,47,48], and we therefore selected the higher dose for this study (~ 0.3 mg/mouse). There was no difference between the body weights of mice treated with vehicle or IAXO-101 within a specific cohort (Additional file 1: Fig. S7). To evaluate the effects of IAXO-101, we measured neuroinflammation (astrogliosis, microgliosis, glial cell morphology), Aβ pathology, and behavior in EFAD mice.

PVT paradigm: IAXO-101 lowers neuroinflammation and improves memory in female E4FAD mice

We first focused on whether IAXO-101 could modulate neuroinflammation Aβ pathology and behavior in female E4FAD mice in a prevention paradigm (PVT). Evidence suggests that there is a synergistic effect of APOE4 and female sex on both AD risk and pathology in humans [49,50,51,52] and neuroinflammation in vivo [37, 41, 53,54,55]. Indeed, female E4FAD mice have higher neuroinflammation and Aβ pathology compared to male E4FAD mice at 6 months of age [15, 19, 56]. Therefore, we considered female E4FAD mice were a logical starting point to test if TLR4 plays a role in regulating neuroinflammation.

In the brain, glia (astrocytes and microglia) are key components of the neuroinflammatory response. Alterations in astrocyte reactivity can be evaluated via quantification of GFAP (Fig. 1A–C), S100β [57], and C3 [58] (Fig. S2A-B), which we measured in female E4FAD mice by IHC analysis. We did not find any differences in the levels of any of those astrocytic markers between vehicle and IAXO-101-treated mice. Microglia reactivity can be measured through quantification of Iba-1 (Fig. 1D) and Clec7a [59] levels (IHC). We found that IAXO-101 treatment resulted in a non-significant trend of lower Iba-1 coverage levels in the CX (Fig. 1D–E), and ~ 30% lower levels in the HP (Fig. 1F). There was also a non-significant trend of lower Clec7a coverage in the CX and HP with IAXO-101 treatment (Figure S2C). As cortical Iba-1 was trending lower with IAXO-101, we further evaluated microglial reactivity by calculating the total number in eight sub-regions of the CX (Fig. 1G). We found that total number of microglia (Iba-1+) was lower number with IAXO-101 treatment (Fig. 1H). Microglia states can be classified morphologically as resting (small soma, thin processes; type 1) or reactive (amoeboid; type 2/3), the latter of which is associated with pathological conditions. We found that with IAXO-101 treatment, the number of resting microglia did not change (Fig. 1I), but the number of reactive microglia was lower (Fig. 1J). Further analysis revealed that the distribution of microglial subtypes was altered by IAXO-101 treatment, with a ~ 5% decrease in type 2/3 microglia in IAXO-101 treated mice (Fig. 1K). Changes in microglia reactivity are often associated with altered cytokine levels, including IL-1β. Consistent with lower Iba-1 coverage, IL1-β levels were ~ 25% lower in the CX and ~ 60% lower in the HP of IAXO-101-treated female E4FAD mice (Fig. 1L, M).

Fig. 1
figure 1

PVT paradigm: IAXO-101 lowers neuroinflammation and improves memory in female E4FAD mice. Female E4FAD mice were treated with IAXO-101 or vehicle from 4 to 6 months of age; PVT paradigm. A Representative image of mouse brains immunostained for GFAP (green, scale bars: 1000 μm). Treatment did not impact the percentage of area covered by GFAP in the B cortex (CX) [t(13.46) = 0.5080, p > 0.5] or the C hippocampus (HP) [t(13.79) = 0.1996, p > 0.5]. D Microglia Iba-1 coverage (green, scale bars: 1000 μm). IAXO-101 treatment appeared to lower the percentage area covered by Iba-1 in the E CX [t(7.998) = 1.909, p = 0.093] and the F HP [t(7.259) = 2.608, p < 0.05]. G Higher power magnification images of Iba-1 in the CX (green, scale bars: 50 μm). IAXO-101 treatment H resulted in a lower number of total microglia [t(9.809) = 0.5045, p = 0.05], and I did not affect type 1 microglia; however, J resulted in a lower number of type 2/3 microglia. K Percentage of type 1 and 2/3 microglia [type 1: t(10.81) = 1.456, p > 0.1; type 2/3: t(10.03) = 0.5080, p < 0.05]. Levels of IL-1β were lower with IAXO-101 treatment in the L CX [t(9) = 2.297, p < 0.05] and M the HP [t(9) = 2.499, p < 0.05]. N Representative images of Aβ immunostaining (red, scale bars: 1000 μm). There was no effect of IAXO-101 on Aβ levels in the O CX [t(8.644) = 1.680, p > 0.1] and the P HP [t(13.86) = 1.031, p > 0.1]. IAXO-101 treatment increased fibrillar amyloid (Q green, scale bars: 1000 μm) in the R CX [t(6.953) = 2.608, p < 0.05] with no effect in the S HP [t(11.47) = 1.317, p > 0.1]. In the Morris water maze test, IAXO-101 treatment T had no effect on the learning/acquisition [2-way ANOVA-days: F(3.111,40.45) = 11.93, p < 0.0001; treatment: F(1,13) = 1.011, p = 0.333]; however, U in the memory/probe trial resulted in lower latency to quadrant and platform [target quadrant: t(7.513) = 2.528, p < 0.05; platform: t(6.604) = 2.388, p = 0.05. Data are expressed as mean ± S.E.M. All data analyzed by Student’s t-test, except in Morris water maze acquisition trials (two-way repeated measures ANOVA). * p < 0.05. See Additional file 3 for n sizes and statistical analysis

One of the proposed functions of glia in AD, particularly microglia, is to clear Aβ [60, 61]. Therefore, Aβ42 levels could have been impacted by IAXO-101 treatment, which we measured using biochemical analysis (BC, ELISA). Surprisingly, there was no effect of IAXO-101 treatment on soluble and insoluble A β42 levels in the CX and HP (BC; Fig. S3A, B). To confirm that IAXO-101 treatment did not modulate extracellular Aβ, we performed immunohistochemical (IHC) analysis (MOAB-2) for Aβ (Fig. 1N) and Thio-S staining for fibrillar amyloid deposits (Fig. 1Q) in female E4FAD mice. IAXO-101 treatment did not impact Aβ deposition (Fig. 1 O, P) or fibrillar amyloid deposits in HP (Fig. 1S) but increased fibrillar amyloid deposits in the CX (Fig. 1R). We also found that soluble and insoluble apoE levels were similar between vehicle- and IAXO-101-treated mice (Fig. S3C, D).

Since IAXO-101 lowered markers of neuroinflammation, we next evaluated the potential impact on learning/memory-relevant using Morris water maze. In the acquisition phase, both groups of mice learned the location of the platform, with no differences between treatments (Fig. 1T). However, in probe trial (memory), IAXO-101-treated female E4FAD mice had lower latency to the target quadrant and platform (Fig. 1U).

In summary, IAXO-101 lowered microglial reactivity and IL-1β levels and improved memory in female E4FAD mice treated in PVT paradigm.

RVS paradigm: IAXO-101 lowers neuroinflammation and Aβ pathology and improves memory in female E4FAD mice

An important component of AD research is understanding the extent that targeting a particular function or pathway can modulate pathology and cognition at advanced stages of the disease. Therefore, we next determined the impact of TLR4 antagonism on neuroinflammation and behavior at an age of significant Aβ pathology [19], by treating female E4FAD from 6 to 7 months of age with either vehicle or IAXO-101. Although not significant, GFAP (Fig. 2A–C) and S100β (Fig. S4A) coverage was trending lower with IAXO-101 treatment, with no effect on C3 (Fig. S4B). IAXO-101 lowered Iba-1+ microglia coverage by ~ 40% in the CX (Fig. 2D, E) and ~ 15% in the HP (Fig. 2D, F). Consistent with this observation, we found that IAXO-101 also lowered Clec7a + microglia coverage by 20% in the CX (p = 0.09) and HP (Fig. S4C). Subsequent morphological analysis revealed that IAXO-101 treatment was associated with lower number of total number of microglia (Fig. 2H, J), no change in the number of resting microglia (Fig. 2I), and a lower number of reactive microglia (Fig. 2J) and ~ 5% lower distribution of type 2/3 microglia (Fig. 2K). Surprisingly, we did not detect any changes in IL1-β levels in female E4FAD mice (Fig. 2L, M). IAXO-101 treatment also resulted in lower levels of Aβ plaques (Fig. 2N–P) and fibrillar amyloid deposits (Fig. 2Q–S), in both the CX and the HP, but no changes in Aβ (Fig. S3E, F) or apoE levels by BC analysis (Fig. S2G, H). At the behavioral level, IAXO-101 improved memory in the Morris water maze (Fig. 2T, U). Overall, in a RVS paradigm, IAXO-101 treatment lowered neuroinflammation and Aβ pathology and improved memory in female E4FAD mice.

Fig. 2
figure 2

RVS paradigm: IAXO-101 lowers neuroinflammation and Aβ pathology and improves memory in female E4FAD mice. Female E4FAD mice were treated with IAXO-101 or vehicle from 6 to 7 months of age; RVS paradigm. A Representative image of mouse brains immunostained for GFAP (green, scale bars: 1000 μm). IAXO-101 treatment did not affect the percentage of area covered by GFAP in the B cortex (CX) [t(13.71) = 0.9136, p > 0.5] or the C hippocampus (HP) [t(9.278) = 1.448, p > 0.5]. D Microglia Iba-1 coverage (green, scale bars: 1000 μm). IAXO-101 treatment lowered Iba-1 coverage in the E CX [t(7.345) = 4.671, p < 0.01] and the F HP [t(8.823) = 2.309, p < 0.05]. G Higher power magnification images of Iba-1 in the CX (green, scale bars: 50 μm). IAXO-101 treatment H resulted in a lower number of total microglia [t(9.995) = 3.870, p < 0.001], and I did not affect type 1 microglia; however, J resulted in a lower number of type 2/3 microglia. K Percentage of type 1 and 2/3 microglia [type 1: t(9.998) = 0.2848, p > 0.1; type 2/3: t(9.793) = 4.073, p < 0.001]. Levels of IL-1β did not change with IAXO-101 treatment in the L CX [t(12.18) = 1.039, p > 0.1] and M the HP [t(7.979) = 0.928, p > 0.1]. N Representative images of Aβ immunostaining (red, scale bars: 1000 μm). IAXO-101 treatment lowered Aβ levels in the O CX [t(13.53) = 2.040, p = 0.06] and the P HP [t(11.46) = 2.294, p < 0.05]. IAXO-101 treatment lowered fibrillar amyloid (Q green, scale bars: 1000 μm) in the R CX [t(10.13) = 2.056, p = 0.06] and in the S HP [t(8.520) = 3.725, p < 0.01]. In the Morris water maze test, IAXO-101 treatment T had no effect on the learning/acquisition [2-way ANOVA-days: F(2.907,34.89) = 3.204, p < 0.05; treatment: F(1,12) = 2.807, p = 0.1197]; however, U in the memory/probe trial resulted in lower latency to quadrant and platform [target quadrant: t(8.663) = 1.7, p = 0.1248; platform: t(8.991) = 2.462, p < 0.05]. Data are expressed as mean ± S.E.M. All data analyzed by Student’s t-test, except in Morris water maze acquisition trials T (two-way repeated measures ANOVA). * p < 0.05. See Additional file 3 for n sizes and statistical analysis

PVT paradigm: IAXO-101 has no effect on specific neuroinflammatory and behavioral readouts in male E4FAD mice

In general, data supports that the combination of female sex and APOE4 induces a strong neuroinflammatory phenotype in FAD-mice. However, male E4FAD mice also have greater neuroinflammation and Aβ pathology compared to male E3FAD mice at 6 months of age [11, 15, 19, 35]. Therefore, we evaluated whether the beneficial effect of IAXO-101 in lowering neuroinflammation also applied to male E4FAD mice in the same PVT paradigm (4 to 6 months treatment). There was no effect of IAXO-101 treatment on neuroinflammatory markers including GFAP (Fig. 3A–C) and Iba-1 coverage (Fig. 3D–F), total number of microglia (Fig. 3G, H), number of resting (Fig. 3I) and reactive microglia (Fig. 3J), distribution of microglial subtypes (Fig. 3K), and IL1-β levels (Fig. 3L, M). We also did not detect any changes in fibrillar amyloid or Aβ levels (Fig. 3N–S, Fig. S2I, J), apoE levels (Fig. S3K, L), or learning/memory (Fig. 3T, U) after IAXO-101 treatment. Thus, in contrast to female E4FAD mice, IAXO-101 treatment did not result in any beneficial effects on our readouts in male E4FAD mice in a PVT paradigm.

Fig. 3
figure 3

PVT paradigm: IAXO-101 has no effect on specific neuroinflammatory and behavioral readouts in male E4FAD mice. Male E4FAD mice were treated with IAXO-101 or vehicle from 4 to 6 months of age; PVT paradigm. A Representative image of mouse brains immunostained for GFAP (green, scale bars: 1000 μm). Treatment did not impact the percentage of area covered by GFAP in the B cortex (CX) [t(10.68) = 0.1806, p > 0.5] or the C hippocampus (HP) [t(9.084) = 1.503, p > 0.1]. D Microglia Iba-1 coverage (green, scale bars: 1000 μm). IAXO-101 treatment had no effect on percentage area covered by Iba-1 in the E CX [t(9.765) = 0.9540, p > 0.1] and the F HP [t(9.829) = 0.3946, p > 0.5]. G Higher power magnification images of Iba-1 in the CX (green, scale bars: 50 μm). IAXO-101 treatment H had no effect on number of total microglia I type 1 microglia, J type 2/3 microglia, K distribution of microglial subtypes [total: t(11.98) = 1.226, p > 0.1], type 1: t(10.73) = 0.09, p > 0.5; type 2/3: t(10.86) = 1.567, p > 0.1] or levels of IL-1β in the L CX [t(11) = 0.6826, p < 0.5] and M the HP [t(12) = 1.095, p > 0.1]. N Representative images of Aβ immunostaining (red, scale bars: 1000 μm). There was no effect of IAXO-101 on Aβ levels in the O CX [t(11.38) = 1.171, p > 0.1] and the P HP [t(9.549) = 0.7235, p > 0.1] or in fibrillar amyloid (Q green, scale bars: 1000 μm) in the R CX [t(11.45) = 1.07, p > 0.1] and the S HP [t(11.69) = 1.543, p > 0.1]. In the Morris water maze test, IAXO-101 treatment T had no effect on the learning/acquisition [2-way ANOVA-days: F(3.266,45.72) = 4.532, p < 0.01; treatment: F(1,14) = 1.056, p = 0.322] and U in the memory/probe trials [target quadrant: t(10.74) = 0.9724, p > 0.1; platform: t(11.48) = 0.004, p > 0.5]. Data are expressed as mean ± S.E.M. All data analyzed by Student’s t-test, except in Morris water maze acquisition trials (two-way repeated measures ANOVA). * p < 0.05. See Additional file 3 for n sizes and statistical analysis

RVS paradigm: IAXO-101 treatment lowered Iba-1 coverage but had no effect on Aβ pathology and behavior in male E4FAD mice

One possible explanation for a lack of an effect of IAXO-101 in PVT paradigms in male E4FAD mice is that at early stages/ages of pathology the contribution of TLR4 to neuroinflammation and/or behavioral function is minimal. To explore this concept, we next evaluated the effect of TLR4 antagonism on behavior in male E4FAD mice at ages of more advanced Aβ levels and higher neuroinflammation. There are age-dependent increases in Aβ pathology and neuroinflammation in male E4FAD mice from 4 to 6 months of age [35]. Therefore, we next determined whether IAXO-101 treatment from 6 to 7 months of age was beneficial in male E4FAD mice. IAX0-101 did not affect GFAP coverage (Fig. 4A–C). However, IAXO-101 treatment reduced Iba-1 coverage by ~ 50% the CX (Fig. 4D, E) and ~ 25% in the HP (Fig. 4D, F). In addition, compared to vehicle, IAXO-101 treatment resulted in a lower number of total microglia (Fig. 4G, H) and lower percentage of reactive microglial subtypes (Fig. 4I–K) without modulating IL-1β levels (Fig. 4L, M). There was no effect of IAXO-101 on Aβ levels (Fig. S3O, P, Fig. 4N-S), apoE levels (Fig. S3M, N), or learning and memory (Fig. 4T, U). Thus, in general, the effect of IAXO-101 in male E4FAD mice may be limited to lowering neuroinflammatory markers without affecting Aβ pathology or cognition.

Fig. 4
figure 4

RVS paradigm: IAXO-101 treatment lowered Iba-1 coverage but had no effect on Aβ pathology and behavior in male E4FAD mice. Male E4FAD mice were treated with IAXO-101 or vehicle from 6 to 7 months of age; RVS paradigm. A Representative image of mouse brains immunostained for GFAP (green, scale bars: 1000 μm). Treatment did not impact the percentage of area covered by GFAP in the B cortex (CX) [t(11.16) = 0.6788, p > 0.5] or the C hippocampus (HP) [t(13.87) = 0.1201, p > 0.5]. D Microglia Iba-1 coverage (green, scale bars: 1000 μm). IAXO-101 treatment lowered percentage area covered by Iba-1 in the E CX [t(8.742) = 4.186, p < 0.05] and the F HP [t(9.993) = 2.253, p < 0.5]. G Higher power magnification images of Iba-1 in the CX (green, scale bars: 50 μm). IAXO-101 treatment H lowered number of total microglia I but did not affect type 1 microglia [total: t(9.035) = 2.497, p < 0.05; type 1: t(10.00) = 0.3145, p > 0.5]. However, IAXO-101 treatment lowered J type 2/3 microglia [type 2/3: t(8.845) = 4.032, p > 0.1] and modified the K distribution of microglial subtypes. Surprisingly, there was no effect of IAXO-101 on the levels of IL-1β in the L CX [t(9.835) = 0.177, p > 0.5] and M the HP [t(5.356) = 1.348 p > 0.1]. N Representative images of Aβ immunostaining (red, scale bars: 1000 μm). There was no effect of IAXO-101 on Aβ levels in the O CX [t(7.636) = 0.8443, p > 0.1] and the P HP [t(12.63) = 1.643, p > 0.1] or in fibrillar amyloid (Q green, scale bars: 1000 μm) in the R CX [t(9.761) = 10.558, p > 0.5] and the S HP [t(13.47) = 0.2563, p > 0.5]. In the Morris water maze test, IAXO-101 treatment T had no effect on the learning/acquisition [2-way ANOVA-days: F(2.774,38.84) = 6.170, p < 0.01; treatment: F(1,14) = 0.064, p > 0.5] and U in the memory/probe trials [target quadrant: t(8.075) = 0.035, p > 0.5; platform t(7.641) = 1.134, p > 0.1]. Data are expressed as mean ± S.E.M. All data analyzed by Student’s t-test, except in Morris water maze acquisition trials (two-way repeated measures ANOVA). * p < 0.05. See Additional file 3 for n sizes and statistical analysis

PVT/RVS paradigm: IAXO-101 had no effect in female E3FAD mice

Our data suggested that IAXO-101 is more beneficial in female E4FAD mice than males, particularly for microglia, which raised the question of whether TLR4 antagonism would be beneficial for female APOE3 carriers. Indeed, neuroinflammation, including microglial number and morphology, has been shown to be higher in females in rodents [62] and humans [63]. Therefore, we tested if IAXO-101 affected the levels of neuroinflammation in female E3FAD mice in both PVT and RVS paradigms. We found that IAXO-101 did not affect neuroinflammation (PVT: Fig. S5A-F; RVS: Fig. S6A-F), Aβ or apoE levels (PVT: Fig. S5G-J; RVS: Fig. S6G-J) or learning and memory (PVT: Fig. S5K, L; RVS: Fig. S6K, L) in female E3FAD mice. Therefore, unlike female APOE4 mice, IAXO-101 was not effective at mitigating pathology and behavior in female E3FAD mice.

Discussion

Neuroinflammation mediated by microglia is increasingly recognized as an important component of AD pathogenesis. However, the role of microglia function in AD is complex and is dependent on many factors including activation state and disease stage. For example, it has been proposed that moderately activated microglia are neuroprotective in the early stages of AD, while chronically activated microglia may be detrimental in later stages (reviewed in [64,65,66]). Genetic AD risk factors and sex may also impact the extent that microglia are beneficial, neutral, or detrimental for cognitive function in AD. Our study supports that reducing microglial neuroinflammation is particularly or specifically beneficial for improving memory in female APOE4 carriers. Although there are limitations (see below), the implication is that neuroinflammation may contribute to higher risk and progression of AD found in APOE4 females. Consistent with this concept, human data demonstrate that compared to APOE3, female APOE4 AD patients have earlier onset of cognitive deficits (reviewed in [67]) and increased neuroinflammation, including increased activation of microglia and secretion of cytokines [7,8,9,10]. Similarly, in mice, greater impairment in learning/memory and neuroinflammation is seen in females compared to males in multiple FAD mouse models [68,69,70,71,72], with APOE4 exacerbating these effects [11, 15, 19]. Thus, collectively, the combination of female sex and APOE4 may produce an environment that specifically results in greater microglial inflammatory respond that leads to greater cognitive dysfunction in AD.

The concept that APOE4 and female sex synergistically impact neuroinflammation to impair cognition raises the important discussion of the underlying mechanisms. In general, apoE4 has been proposed to alter cellular function in different ways [73]. At the structural level, apoE isoforms differ by two amino acids at position 112 and 158, which affects protein folding and is thought to result in changes to lipidation (apoE4 < apoE3) stability and levels (apoE4 < apoE3) [74, 75]. These structural changes are proposed to induce differences apoE availability, distribution, aggregation, receptor binding affinities and signaling that modulates peripheral inflammation (via macrophages and other immune cells), cerebrovascular function (via endothelial cells, pericytes and astrocytes), Aβ levels, lipid trafficking, and neuron function directly (reviewed in [3, 76,77,78,79]). Therefore, the impact of APOE on other cell types could indirectly impact glial morphology/activation. In addition, apoE can be produced by microglia to impact their activity. For example, microglia expressing APOE4 has altered immune responses and metabolism in vitro [18, 80, 81], and selective ablation of microglial apoE4 in a tauopathy mouse model blocks brain atrophy [82, 83]. Thus, collectively APOE4 could indirectly and directly results in higher potential for microglia reactivity to be exacerbated by female sex.

The underlying mechanisms of how female sex impacts AD progression is unclear. One proposed mechanism is that loss of sex hormones at menopause results in greater AD susceptibility. Indeed, ovariectomy results in behavioral impairments and accelerated aging [84,85,86] in women as well as in rodents [87, 88]. Importantly, ovariectomy results in greater neuroinflammation in rodents including microglial activation [87,88,89,90,91]. Therefore, as estrogen demonstrates anti-inflammatory activity via the estrogen receptors, the loss of estrogen may promote a microglial response [92]. In addition to direct effects on microglia, the loss of estrogen also modulates AD pathology, as in general, Aβ deposition is increased in women during the menopausal transition [93, 94], in post-mortem brains of ovariectomized women [95], and in brains of ovariectomized rodents with some conflicting results in FAD mice (discussed in [96]). Thus, menopause could indirectly and directly modulate microglial activation in a way that is particularly pronounced with APOE4 carriers. However, in our study, mice were not ovariectomized, suggesting a different mechanism of action for how female sex and APOE4 impacted glial activation. In general, there are sex differences in microglial numbers, function, and response to an acute/chronic insult and age without menopause mimics [62, 97], including in APOE4 FAD mice [11, 34]. Thus, one potential explanation is with APOE4, there is greater age-dependent changes in sex hormones or their receptors that influence the inflammatory state of microglia, thereby modulating its function. Alternatively, the difference between sex hormones and immune-related genes on the X chromosome may result in higher age-related neuroinflammation (reviewed in [98,99,100,101]). Future studies could focus on understanding in more detail the precise mechanism(s) though which sex and APOE4 increase neuroinflammation and microglial activation.

Our data supports a specific role of TLR4 in modulating neuroinflammation and cognition in APOE4 females. Data from human, in vivo and mouse studies have led to the idea that blocking TLR4 is a potential AD therapeutic strategy albeit in a non-APOE4 context. Several single nucleotide polymorphisms (SNPs) in TLR4 have been associated with modulating AD risk in humans. For example, the G coding variant of rs4986790(A/G) [31, 32] and the C variant of rs11367(G/C) in TLR4 is associated with decreased AD risk [102]. Consistent with a detrimental role of TLR4 in AD, in vitro and ex vivo studies supports that these variants reduce TLR4 activation, resulting in attenuation of proinflammatory signaling [103, 104]. Several minor alleles of TLR4 SNPs (rs10759930, rs1927914, rs1927911, rs12377632) were also found to increase the risk of AD [33], although their function is yet to be understood. Further evidence for a role of TLR4 in AD include that levels of LPS [105] and TLR4 expression are increased in the brains of AD patients [28, 29] and FAD mice [29, 30]. In addition, Aβ has been shown to activate TLR4 resulting in reactive oxygen species production in microglia in vitro [106], and deletion of the TLR4 co-receptor CD14 resulted in lower amyloid plaques in vivo [107]. An important question for further study is whether SNPs in TLR4, TLR4 expression data, and in vitro functional data are modulated by APOE and sex in AD-relevant context. In terms of APOE alone, LPS (TLR4 agonist) induces APOE-specific neuroinflammatory markers in multiple tissues (APOE4 > APOE3) in vivo [14, 108], ex vivo [108], and in vitro [109] with an inconsistent sex effect [110]. In terms of female sex, TLR4 expression is greater in multiple tissues in females than males in human and rodents [110], and the estrogen receptor may impact TLR signaling [92]. Potentially, therefore, the combination of APOE and female sex may specifically result in altered TLR4-mediated neuroinflammation to impact cognition.

The implication of our findings is that targeting neuroinflammation is a potential AD therapeutic approach; for certain high-risk groups, however, current human studies are conflicted. To date, most data is derived from epidemiological studies, and a few clinical trials focused on established drugs with known anti-inflammatory properties. One class is NSAIDs that inhibit COX1 and 2 activities. Epidemiological and in vivo studies indicated that non-steroidal anti-inflammatory drugs (NSAIDs) lower AD risk and pathology (reviewed in [111, 112]). However, NSAIDs have not been found efficacious in some AD clinical trials [20]. In addition, a recent re-analysis of the NSAID epidemiological data in an ADNI dataset revealed that although some NSAIDs are associated with decreased AD prevalence, they did not affect cognitive decline in AD patients [21]. A second drug used to evaluate neuroinflammation as an AD target is paracetamol that has unknown and likely pleiotropic mechanisms of action. For example, paracetamol is used for its anti-pyretic, anti-inflammatory, and analgesic actions that may be mediated through lowering prostaglandin levels, inhibiting COX, altering cannabinoid signaling, and also blocking neuronal sodium channels [113]. Paracetamol has been shown to lower AD prevalence; however, whether the beneficial effects of paracetamol in AD are due to effects on inflammation or other mechanisms is unclear [21]. In addition to further understanding the mechanism of action of anti-inflammatories, there are several important considerations for clinically targeting neuroinflammation in AD. One is that neuroinflammation is complex and recognition of pathogens or danger-associated molecules occurs in multiple cell types, through a large array of receptors that produce an equally complex signaling and functional response. Thus, targeted one specific pathway may not alone be sufficient to improve all aspects of neuroinflammation in AD. Therefore, a detailed understanding of how different pathways contribute to any proposed detrimental neuroinflammatory phenotype in AD, including which aspects are more proximal to neuronal dysfunction, is important for interpreting clinical trial data. A second is that whether an inflammatory mediator or receptor is beneficial or detrimental for AD progression may depend on the stage of disease. For example, it has proposed that anti-inflammatories may be efficacious as a prophylactic treatment for APOE4 carriers to prevent AD [21]. A third, as highlighted by our study, is that the contribution of neuroinflammation to neural dysfunction may be dependent on risk factors (e.g., sex, APOE genotype, lifestyle, other genetic or environmental factors). Perhaps future studies in larger heterogenous populations could provide an explanation for determining whether NSAIDs have a greater impact in specific groups.

Limitations

There are limitations in the extent that we can conclude TLR4 and/or neuroinflammation specifically contributes to neural dysfunction in female APOE4 mice. In fact, studies have shown that microglial activation and neuroinflammation are greater with APOE4 than APOE3 in males using APOE knock in mice [14, 114,115,116], FAD mice [11, 15, 43], and AD patients [117, 118]. Indeed, it is important to note that we found that IAXO-101 lowered microglial number and reactivity in older male E4FAD mice, although performance in the Morris water maze test was not affected. The contribution of TLR4 to microglial activation maybe dependent on age and/or Aβ pathology, which is why IAXO-101 modulated microglia number/reactivity when treatment occurred in older rather than younger male E4FAD mice. The lack of a behavioral benefit in male E4FAD mice as opposed to female E4FAD mice with IAXO-101 treatment could be due multifactorial factors. For example, there could be the same age-dependent consideration as for microglial activation within males, such that beneficial effects in behavior would have been observed if treatment had continued for longer. Alternatively, the contribution of microglia to performance in Morris water maze performance may be greater in females than males. Furthermore, a more comprehensive set of behavioral tests along with expanded markers of neuron function may have revealed that IAXO-101 was beneficial in E4FAD male mice. In addition, APOE3-FAD also shows age-dependent increases in glial activation and cognitive impairment [19, 40]. Therefore, future studies could explore if TLR4, neuroinflammation, and/or microglia activity are more proximal to behavioral dysfunction in older male E4FAD, female E3FAD, and male E3FAD mice. Such studies could reveal if there are different optimal treatment windows for targeting neuroinflammation within each APOE genotype and sex combination or support specificity for female APOE4 carriers.

Our study was a proof-of-concept design centered on TLR4 antagonism. However, TLR4 may not be the main contribution to inflammation for all APOE genotypes and sexes. For example, there is the possibility that TLR4 activation is more pronounced in female APOE4 mice, but other inflammatory pathways are important for APOE4 male mice or APOE3 mice. Indeed, sex hormones and X chromosome genes can differentially regulate TLR4 [98, 99, 119] and testosterone decreases expression and function of pro-inflammatory cytokines [120]. In addition, as mentioned above, different inflammatory pathways may be important at different ages/stages of pathology. Therefore, identifying how APOE genotype, sex, and age impact the neuroinflammatory phenotype, including specific pathways, is important for ultimately advancing our mechanistic understanding of neuroinflammation in AD.

There were also experimental limitations surrounding the way that we targeted TLR4. One aspect is the dose of IAXO-101, which we selected based on previous publications; however, we did not conduct detailed PK studies to determine brain and plasma levels. Therefore, IAXO-101 levels in the plasma and brain may have been different for each group of mice, such as higher in female E4FAD mice, which may explain the beneficial effects. In addition, IAXO-101 inhibits TLR4 activation by two mechanisms: sequestering LPS by forming stable co-aggregates or competing with LPS for binding to CD14 and myeloid differentiation factor 2 [121]. This mechanism of action is based on LPS; however, it is unclear what pathways lead to TLR4 activation in female E4FAD mice. Thus, another approach would be to use competitive inhibitors of TLR4 and/inhibitors of TLR4 signaling down-stream. There is also the question of whether there are cell type specific functions of TLR4. TLR4 is expressed by virtually every cell in the body that could have opposed or synergistic functions. In fact, both activating and inhibiting TLR4 has been shown to decrease AD-related pathology in FAD mice with opposing effects on neuroinflammation [122, 123]. Therefore, evaluating the cell type-specific functions of TLR4 in EFAD mice using detailed mechanistic readouts for neuroinflammation, neuron function, and behavior as well as plasma biomarkers is important for fully evaluating TLR4 as a therapeutic target in AD.

Conclusion

Our study demonstrates that the TLR4 antagonist IAXO-101 improves memory and reduced neuroinflammation with minimal effects of Aβ pathology in female APOE4 mice. Thus, TLR4 inhibition is a potential therapeutic approach for AD, particularly in female APOE4 carriers. In addition, these results support stratification of preclinical and clinical studies that target neuroinflammation by APOE, sex, and treatment window.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

Aβ:

Amyloid beta

AD:

Alzheimer’s disease

Apolipoprotein E:

ApoE (gene: APOE)

CD14:

Cluster of differentiation 14

COX:

Cyclooxygenase

CX:

Cortex

FAD:

Familial Alzheimer’s disease

IL:

Interleukin

LPS:

Lipopolysaccharide

NSAID:

Non-steroidal anti-inflammatory drugs

PVT:

Prevention

RVS:

Reversal

SNP:

Single nucleotide polymorphism

TLR:

Toll-like receptor

References

  1. Rahman A, Jackson H, Hristov H, Isaacson RS, Saif N, Shetty T, et al. Sex and gender driven modifiers of Alzheimer’s: the role for estrogenic control across age, race, medical, and lifestyle risks. Front Aging Neurosci. 2019;11:315.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Riedel BC, Thompson PM, Brinton RD. Age, APOE and sex: triad of risk of Alzheimer’s disease. J Steroid Biochem Mol Biol. 2016;160:134–47.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Raulin AC, Doss SV, Trottier ZA, Ikezu TC, Bu G, Liu CC. ApoE in Alzheimer’s disease: pathophysiology and therapeutic strategies. Mol Neurodegener. 2022;17(1):72.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Rueter J, Rimbach G, Huebbe P. Functional diversity of apolipoprotein E: from subcellular localization to mitochondrial function. Cell Mol Life Sci. 2022;79(9):499.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Tai LM, Thomas R, Marottoli FM, Koster KP, Kanekiyo T, Morris AW, et al. The role of APOE in cerebrovascular dysfunction. Acta Neuropathol. 2016;131(5):709–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Parhizkar S, Holtzman DM. APOE mediated neuroinflammation and neurodegeneration in Alzheimer’s disease. Semin Immunol. 2022;59:101594.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Friedberg JS, Aytan N, Cherry JD, Xia W, Standring OJ, Alvarez VE, et al. Associations between brain inflammatory profiles and human neuropathology are altered based on apolipoprotein E epsilon4 genotype. Sci Rep. 2020;10(1):2924.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Reale M, Kamal MA, Velluto L, Gambi D, Di Nicola M, Greig NH. Relationship between inflammatory mediators, Abeta levels and ApoE genotype in Alzheimer disease. Curr Alzheimer Res. 2012;9(4):447–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Ringman JM, Elashoff D, Geschwind DH, Welsh BT, Gylys KH, Lee C, et al. Plasma signaling proteins in persons at genetic risk for Alzheimer disease: influence of APOE genotype. Arch Neurol. 2012;69(6):757–64.

    PubMed  PubMed Central  Google Scholar 

  10. Tzioras M, Davies C, Newman A, Jackson R, Spires-Jones T. Invited review: APOE at the interface of inflammation, neurodegeneration and pathological protein spread in Alzheimer’s disease. Neuropathol Appl Neurobiol. 2019;45(4):327–46.

    CAS  PubMed  Google Scholar 

  11. Stephen TL, Cacciottolo M, Balu D, Morgan TE, LaDu MJ, Finch CE, et al. APOE genotype and sex affect microglial interactions with plaques in Alzheimer’s disease mice. Acta Neuropathol Commun. 2019;7(1):82.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Rodriguez GA, Tai LM, LaDu MJ, Rebeck GW. Human APOE4 increases microglia reactivity at Abeta plaques in a mouse model of Abeta deposition. J Neuroinflammation. 2014;11(1):111.

    PubMed  PubMed Central  Google Scholar 

  13. Liu CC, Zhao N, Fu Y, Wang N, Linares C, Tsai CW, et al. ApoE4 accelerates early seeding of amyloid pathology. Neuron. 2017;96(5):1024-32 e3.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhu Y, Nwabuisi-Heath E, Dumanis SB, Tai LM, Yu C, Rebeck GW, et al. APOE genotype alters glial activation and loss of synaptic markers in mice. Glia. 2012;60(4):559–69.

    PubMed  PubMed Central  Google Scholar 

  15. Stephen TL, Breningstall B, Suresh S, McGill CJ, Pike CJ. APOE genotype and biological sex regulate astroglial interactions with amyloid plaques in Alzheimer’s disease mice. J Neuroinflammation. 2022;19(1):286.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Vitek MP, Brown CM, Colton CA. APOE genotype-specific differences in the innate immune response. Neurobiol Aging. 2009;30(9):1350–60.

    CAS  PubMed  Google Scholar 

  17. Vitek MP, Christensen DJ, Wilcock D, Davis J, Van Nostrand WE, Li FQ, et al. APOE-mimetic peptides reduce behavioral deficits, plaques and tangles in Alzheimer’s disease transgenics. Neurodegener Dis. 2012;10(1–4):122–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Moser VA, Workman MJ, Hurwitz SJ, Lipman RM, Pike CJ, Svendsen CN. Microglial transcription profiles in mouse and human are driven by APOE4 and sex. iScience. 2021;24(11):103238.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Balu D, Karstens AJ, Loukenas E, Maldonado Weng J, York JM, Valencia-Olvera AC, et al. The role of APOE in transgenic mouse models of AD. Neurosci Lett. 2019;707:134285.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Melchiorri D, Merlo S, Micallef B, Borg JJ, Drafi F. Alzheimer’s disease and neuroinflammation: will new drugs in clinical trials pave the way to a multi-target therapy? Front Pharmacol. 2023;14:1196413.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Rivers-Auty J, Mather AE, Peters R, Lawrence CB, Brough D. Anti-inflammatories in Alzheimer’s disease-potential therapy or spurious correlate? Brain Commun. 2020;2(2):fcaa109.

    PubMed  Google Scholar 

  22. Hampel H, Caraci F, Cuello AC, Caruso G, Nistico R, Corbo M, et al. A path toward precision medicine for neuroinflammatory mechanisms in Alzheimer’s disease. Front Immunol. 2020;11:456.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Fu WY, Wang X, Ip NY. Targeting neuroinflammation as a therapeutic strategy for Alzheimer’s disease: mechanisms, drug candidates, and new opportunities. ACS Chem Neurosci. 2019;10(2):872–9.

    CAS  PubMed  Google Scholar 

  24. Bruno K, Woller SA, Miller YI, Yaksh TL, Wallace M, Beaton G, et al. Targeting toll-like receptor-4 (TLR4)-an emerging therapeutic target for persistent pain states. Pain. 2018;159(10):1908–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Vaure C, Liu Y. A comparative review of toll-like receptor 4 expression and functionality in different animal species. Front Immunol. 2014;5:316.

    PubMed  PubMed Central  Google Scholar 

  26. Behzadi P, Garcia-Perdomo HA, Karpinski TM. Toll-like receptors: general molecular and structural biology. J Immunol Res. 2021;2021:9914854.

    PubMed  PubMed Central  Google Scholar 

  27. Schaefer L. Complexity of danger: the diverse nature of damage-associated molecular patterns. J Biol Chem. 2014;289(51):35237–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Miron J, Picard C, Frappier J, Dea D, Theroux L, Poirier J. TLR4 Gene expression and pro-inflammatory cytokines in Alzheimer’s disease and in response to hippocampal deafferentation in rodents. J Alzheimers Dis. 2018;63(4):1547–56.

    CAS  PubMed  Google Scholar 

  29. Walter S, Letiembre M, Liu Y, Heine H, Penke B, Hao W, et al. Role of the toll-like receptor 4 in neuroinflammation in Alzheimer’s disease. Cell Physiol Biochem. 2007;20(6):947–56.

    CAS  PubMed  Google Scholar 

  30. Islam R, Rajan R, Choudhary H, Vrionis F, Hanafy KA. Gender differences in Alzheimer’s may be associated with TLR4-LYN expression in damage associated microglia and neuronal phagocytosis. J Cell Physiol. 2022.

  31. Miron J, Picard C, Lafaille-Magnan ME, Savard M, Labonte A, Breitner J, et al. Association of TLR4 with Alzheimer’s disease risk and presymptomatic biomarkers of inflammation. Alzheimers Dement. 2019;15(7):951–60.

    PubMed  Google Scholar 

  32. Balistreri CR, Grimaldi MP, Chiappelli M, Licastro F, Castiglia L, Listi F, et al. Association between the polymorphisms of TLR4 and CD14 genes and Alzheimer’s disease. Curr Pharm Des. 2008;14(26):2672–7.

    CAS  PubMed  Google Scholar 

  33. Chen YC, Yip PK, Huang YL, Sun Y, Wen LL, Chu YM, et al. Sequence variants of toll like receptor 4 and late-onset Alzheimer’s disease. PLoS One. 2012;7(12):e50771.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Tai LM, Ghura S, Koster KP, Liakaite V, Maienschein-Cline M, Kanabar P, et al. APOE-modulated Abeta-induced neuroinflammation in Alzheimer’s disease: current landscape, novel data and future perspective. J Neurochem. 2015;133(4):465–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Youmans KL, Tai LM, Nwabuisi-Heath E, Jungbauer L, Kanekiyo T, Gan M, et al. APOE4-specific changes in Abeta accumulation in a new transgenic mouse model of Alzheimer disease. J Biol Chem. 2012;287(50):41774–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, et al. Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci. 2006;26(40):10129–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Thomas R, Zuchowska P, Morris AW, Marottoli FM, Sunny S, Deaton R, et al. Epidermal growth factor prevents APOE4 and amyloid-beta-induced cognitive and cerebrovascular deficits in female mice. Acta Neuropathol Commun. 2016;4(1):111.

    PubMed  PubMed Central  Google Scholar 

  38. Torgersen ML, Judge PJ, Bada Juarez JF, Pandya AD, Fusser M, Davies CW, et al. Physicochemical characterization, toxicity and in vivo biodistribution studies of a discoidal, lipid-based drug delivery vehicle: Lipodisq nanoparticles containing doxorubicin. J Biomed Nanotechnol. 2020;16(4):419–31.

    CAS  PubMed  Google Scholar 

  39. Huggins C, Pearce S, Peri F, Neumann F, Cockerill G, Pirianov G. A novel small molecule TLR4 antagonist (IAXO-102) negatively regulates non-hematopoietic toll like receptor 4 signalling and inhibits aortic aneurysms development. Atherosclerosis. 2015;242(2):563–70.

    CAS  PubMed  Google Scholar 

  40. Liu DS, Pan XD, Zhang J, Shen H, Collins NC, Cole AM, et al. APOE4 enhances age-dependent decline in cognitive function by down-regulating an NMDA receptor pathway in EFAD-Tg mice. Mol Neurodegener. 2015;10:7.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Thomas R, Morris AWJ, Tai LM. Epidermal growth factor prevents APOE4-induced cognitive and cerebrovascular deficits in female mice. Heliyon. 2017;3(6):e00319.

    PubMed  PubMed Central  Google Scholar 

  42. Valencia-Olvera AC, Balu D, Faulk N, Amiridis A, Wang Y, Pham C, et al. Inhibition of ACAT as a therapeutic target for Alzheimer’s disease is independent of ApoE4 lipidation. Neurotherapeutics. 2023;20(4):1120–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Moser VA, Pike CJ. Obesity accelerates alzheimer-related pathology in APOE4 but not APOE3 mice. eNeuro. 2017;4(3):ENEURO.0077-17.2017.

  44. Youmans KL, Leung S, Zhang J, Maus E, Baysac K, Bu G, et al. Amyloid-beta42 alters apolipoprotein E solubility in brains of mice with five familial AD mutations. J Neurosci Methods. 2011;196(1):51–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Youmans KL, Tai LM, Kanekiyo T, Stine WB Jr, Michon SC, Nwabuisi-Heath E, et al. Intraneuronal Abeta detection in 5xFAD mice by a new Abeta-specific antibody. Mol Neurodegener. 2012;7(1):8.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Toyoda Y, Takada T, Umezawa M, Tomura F, Yamanashi Y, Takeda K, et al. Identification of hepatic NPC1L1 as an NAFLD risk factor evidenced by ezetimibe-mediated steatosis prevention and recovery. FASEB Bioadv. 2019;1(5):283–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Haziak K, Herman AP, Wojtulewicz K, Pawlina B, Paczesna K, Bochenek J, et al. Effect of CD14/TLR4 antagonist on GnRH/LH secretion in ewe during central inflammation induced by intracerebroventricular administration of LPS. J Anim Sci Biotechnol. 2018;9:52.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hermann JK, Ravikumar M, Shoffstall AJ, Ereifej ES, Kovach KM, Chang J, et al. Inhibition of the cluster of differentiation 14 innate immunity pathway with IAXO-101 improves chronic microelectrode performance. J Neural Eng. 2018;15(2):025002.

    PubMed  PubMed Central  Google Scholar 

  49. Altmann A, Tian L, Henderson VW, Greicius MD. Sex modifies the APOE-related risk of developing Alzheimer disease. Ann Neurol. 2014;75(4):563–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ungar L, Altmann A, Greicius MD. Apolipoprotein E, gender, and Alzheimer’s disease: an overlooked, but potent and promising interaction. Brain Imaging Behav. 2014;8(2):262–73.

    PubMed  PubMed Central  Google Scholar 

  51. Therriault J, Benedet AL, Pascoal TA, Mathotaarachchi S, Savard M, Chamoun M, et al. APOEepsilon4 potentiates the relationship between amyloid-beta and tau pathologies. Mol Psychiatry. 2020.

  52. Wang YT, Pascoal TA, Therriault J, Kang MS, Benedet AL, Savard M, et al. Interactive rather than independent effect of APOE and sex potentiates tau deposition in women. Brain Commun. 2021;3(2):fcab126.

    PubMed  PubMed Central  Google Scholar 

  53. Raber J, Wong D, Yu GQ, Buttini M, Mahley RW, Pitas RE, et al. Apolipoprotein E and cognitive performance. Nature. 2000;404(6776):352–4.

    CAS  PubMed  Google Scholar 

  54. Reverte I, Klein AB, Ratner C, Domingo JL, Colomina MT. Behavioral phenotype and BDNF differences related to apoE isoforms and sex in young transgenic mice. Exp Neurol. 2012;237(1):116–25.

    CAS  PubMed  Google Scholar 

  55. Hou X, Adeosun SO, Zhang Q, Barlow B, Brents M, Zheng B, et al. Differential contributions of ApoE4 and female sex to BACE1 activity and expression mediate Abeta deposition and learning and memory in mouse models of Alzheimer’s disease. Front Aging Neurosci. 2015;7:207.

    PubMed  PubMed Central  Google Scholar 

  56. Christensen A, Pike CJ. APOE genotype affects metabolic and Alzheimer-related outcomes induced by Western diet in female EFAD mice. FASEB J. 2018;33(3):4054–66.

  57. Raponi E, Agenes F, Delphin C, Assard N, Baudier J, Legraverend C, et al. S100B expression defines a state in which GFAP-expressing cells lose their neural stem cell potential and acquire a more mature developmental stage. Glia. 2007;55(2):165–77.

    PubMed  PubMed Central  Google Scholar 

  58. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541(7638):481–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Clayton K, Delpech JC, Herron S, Iwahara N, Ericsson M, Saito T, et al. Plaque associated microglia hyper-secrete extracellular vesicles and accelerate tau propagation in a humanized APP mouse model. Mol Neurodegener. 2021;16(1):18.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27(9):1592–9.

    CAS  PubMed  Google Scholar 

  61. Ismail R, Parbo P, Madsen LS, Hansen AK, Hansen KV, Schaldemose JL, et al. The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer’s disease: a longitudinal PET study. J Neuroinflammation. 2020;17(1):151.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Han J, Fan Y, Zhou K, Blomgren K, Harris RA. Uncovering sex differences of rodent microglia. J Neuroinflammation. 2021;18(1):74.

    PubMed  PubMed Central  Google Scholar 

  63. Lynch MA. Exploring sex-related differences in microglia may be a game-changer in precision medicine. Front Aging Neurosci. 2022;14:868448.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17(3):157–72.

    PubMed  Google Scholar 

  65. Cai Y, Liu J, Wang B, Sun M, Yang H. Microglia in the neuroinflammatory pathogenesis of Alzheimer’s disease and related therapeutic targets. Front Immunol. 2022;13:856376.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Gao C, Shen X, Tan Y, Chen S. Pathogenesis, therapeutic strategies and biomarker development based on “omics” analysis related to microglia in Alzheimer’s disease. J Neuroinflammation. 2022;19(1):215.

    PubMed  PubMed Central  Google Scholar 

  67. Emrani S, Arain HA, DeMarshall C, Nuriel T. APOE4 is associated with cognitive and pathological heterogeneity in patients with Alzheimer’s disease: a systematic review. Alzheimers Res Ther. 2020;12(1):141.

    PubMed  PubMed Central  Google Scholar 

  68. Schmid S, Rammes G, Blobner M, Kellermann K, Bratke S, Fendl D, et al. Cognitive decline in Tg2576 mice shows sex-specific differences and correlates with cerebral amyloid-beta. Behav Brain Res. 2019;359:408–17.

    CAS  PubMed  Google Scholar 

  69. Mifflin MA, Winslow W, Surendra L, Tallino S, Vural A, Velazquez R. Sex differences in the IntelliCage and the Morris water maze in the APP/PS1 mouse model of amyloidosis. Neurobiol Aging. 2021;101:130–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Guillot-Sestier MV, Araiz AR, Mela V, Gaban AS, O’Neill E, Joshi L, et al. Microglial metabolism is a pivotal factor in sexual dimorphism in Alzheimer’s disease. Commun Biol. 2021;4(1):711.

    PubMed  PubMed Central  Google Scholar 

  71. Sil A, Erfani A, Lamb N, Copland R, Riedel G, Platt B. Sex differences in behavior and molecular pathology in the 5XFAD model. J Alzheimers Dis. 2022;85(2):755–78.

    PubMed  Google Scholar 

  72. Hou X, Adeosun SO, Zhang Q, Barlow B, Brents M, Zheng B, et al. Differential contributions of ApoE4 and female sex to BACE1 activity and expression mediate A deposition and learning and memory in mouse models of Alzheimer’s disease. Front Aging Neurosci. 2015;7:207.

    PubMed  PubMed Central  Google Scholar 

  73. Fernandez-Calle R, Konings SC, Frontinan-Rubio J, Garcia-Revilla J, Camprubi-Ferrer L, Svensson M, et al. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer’s disease pathology and brain diseases. Mol Neurodegener. 2022;17(1):62.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Mahley RW, Weisgraber KH, Huang Y. Apolipoprotein E: structure determines function, from atherosclerosis to Alzheimer’s disease to AIDS. J Lipid Res. 2009;50(Suppl):S183–8.

    PubMed  PubMed Central  Google Scholar 

  75. Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. Neurobiol Dis. 2014;72 Pt A:3–12.

    PubMed  Google Scholar 

  76. Martinez-Martinez AB, Torres-Perez E, Devanney N, Del Moral R, Johnson LA, Arbones-Mainar JM. Beyond the CNS: The many peripheral roles of APOE. Neurobiol Dis. 2020;138:104809.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Yang LG, March ZM, Stephenson RA, Narayan PS. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol Metab. 2023;34(8):430–45.

    CAS  PubMed  Google Scholar 

  78. Zhang L, Xia Y, Gui Y. Neuronal ApoE4 in Alzheimer’s disease and potential therapeutic targets. Front Aging Neurosci. 2023;15:1199434.

    PubMed  PubMed Central  Google Scholar 

  79. Hunsberger HC, Pinky PD, Smith W, Suppiramaniam V, Reed MN. The role of APOE4 in Alzheimer’s disease: strategies for future therapeutic interventions. Neuronal Signal. 2019;3(2):NS20180203.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Lanfranco MF, Sepulveda J, Kopetsky G, Rebeck GW. Expression and secretion of apoE isoforms in astrocytes and microglia during inflammation. Glia. 2021;69(6):1478–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Lin YT, Seo J, Gao F, Feldman HM, Wen HL, Penney J, et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron. 2018;98(6):1294.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Shi Y, Yamada K, Liddelow SA, Smith ST, Zhao L, Luo W, et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature. 2017;549(7673):523–7.

    PubMed  PubMed Central  Google Scholar 

  83. Shi Y, Manis M, Long J, Wang K, Sullivan PM, Remolina Serrano J, et al. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J Exp Med. 2019;216(11):2546–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Rocca WA, Grossardt BR, Shuster LT. Oophorectomy, menopause, estrogen treatment, and cognitive aging: clinical evidence for a window of opportunity. Brain Res. 2011;1379:188–98.

    CAS  PubMed  Google Scholar 

  85. Jett S, Malviya N, Schelbaum E, Jang G, Jahan E, Clancy K, et al. Endogenous and exogenous estrogen exposures: how women’s reproductive health can drive brain aging and inform Alzheimer’s prevention. Front Aging Neurosci. 2022;14:831807.

    CAS  PubMed  Google Scholar 

  86. Levine ME, Lu AT, Chen BH, Hernandez DG, Singleton AB, Ferrucci L, et al. Menopause accelerates biological aging. Proc Natl Acad Sci U S A. 2016;113(33):9327–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Sanchez K, Wu SL, Kakkar R, Darling JS, Harper CS, Fonken LK. Ovariectomy in mice primes hippocampal microglia to exacerbate behavioral sickness responses. Brain Behav Immun Health. 2023;30:100638.

    PubMed  PubMed Central  Google Scholar 

  88. Ge F, Yang H, Lu W, Shi H, Chen Q, Luo Y, et al. Ovariectomy induces microglial cell activation and inflammatory response in rat prefrontal cortices to accelerate the chronic unpredictable stress-mediated anxiety and depression. Biomed Res Int. 2020;2020:3609758.

    PubMed  PubMed Central  Google Scholar 

  89. Benedusi V, Meda C, Della Torre S, Monteleone G, Vegeto E, Maggi A. A lack of ovarian function increases neuroinflammation in aged mice. Endocrinology. 2012;153(6):2777–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Rana AK, Sharma S, Patial V, Singh D. Lithium therapy subdues neuroinflammation to maintain pyramidal cells arborization and rescues neurobehavioural impairments in ovariectomized rats. Mol Neurobiol. 2022;59(3):1706–23.

    CAS  PubMed  Google Scholar 

  91. Wu B, Song Q, Zhang Y, Wang C, Yang M, Zhang J, et al. Antidepressant activity of omega-3 polyunsaturated fatty acids in ovariectomized rats: role of neuroinflammation and microglial polarization. Lipids Health Dis. 2020;19(1):4.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Villa A, Vegeto E, Poletti A, Maggi A. Estrogens, neuroinflammation, and neurodegeneration. Endocr Rev. 2016;37(4):372–402.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Mosconi L, Rahman A, Diaz I, Wu X, Scheyer O, Hristov HW, et al. Increased Alzheimer’s risk during the menopause transition: a 3-year longitudinal brain imaging study. PLoS One. 2018;13(12):e0207885.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Zeydan B, Tosakulwong N, Schwarz CG, Senjem ML, Gunter JL, Reid RI, et al. Association of bilateral salpingo-oophorectomy before menopause onset with medial temporal lobe neurodegeneration. JAMA Neurol. 2019;76(1):95–100.

    PubMed  Google Scholar 

  95. Bove R, Secor E, Chibnik LB, Barnes LL, Schneider JA, Bennett DA, et al. Age at surgical menopause influences cognitive decline and Alzheimer pathology in older women. Neurology. 2014;82(3):222–9.

    PubMed  PubMed Central  Google Scholar 

  96. Kara F, Belloy ME, Voncken R, Sarwari Z, Garima Y, Anckaerts C, et al. Long-term ovarian hormone deprivation alters functional connectivity, brain neurochemical profile and white matter integrity in the Tg2576 amyloid mouse model of Alzheimer’s disease. Neurobiol Aging. 2021;102:139–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Costa J, Martins S, Ferreira PA, Cardoso AMS, Guedes JR, Peca J, et al. The old guard: age-related changes in microglia and their consequences. Mech Ageing Dev. 2021;197:111512.

    CAS  PubMed  Google Scholar 

  98. Zhao L, Mao Z, Woody SK, Brinton RD. Sex differences in metabolic aging of the brain: insights into female susceptibility to Alzheimer’s disease. Neurobiol Aging. 2016;42:69–79.

    PubMed  PubMed Central  Google Scholar 

  99. Guo L, Zhong MB, Zhang L, Zhang B, Cai D. Sex differences in Alzheimer’s disease: insights from the multiomics landscape. Biol Psychiatry. 2022;91(1):61–71.

    CAS  PubMed  Google Scholar 

  100. Bianchi I, Lleo A, Gershwin ME, Invernizzi P. The X chromosome and immune associated genes. J Autoimmun. 2012;38(2–3):J187–92.

    CAS  PubMed  Google Scholar 

  101. Schurz H, Salie M, Tromp G, Hoal EG, Kinnear CJ, Moller M. The X chromosome and sex-specific effects in infectious disease susceptibility. Hum Genomics. 2019;13(1):2.

    PubMed  PubMed Central  Google Scholar 

  102. Wang LZ, Yu JT, Miao D, Wu ZC, Zong Y, Wen CQ, et al. Genetic association of TLR4/11367 polymorphism with late-onset Alzheimer’s disease in a Han Chinese population. Brain Res. 2011;1381:202–7.

    CAS  PubMed  Google Scholar 

  103. Arbour NC, Lorenz E, Schutte BC, Zabner J, Kline JN, Jones M, et al. TLR4 mutations are associated with endotoxin hyporesponsiveness in humans. Nat Genet. 2000;25(2):187–91.

    CAS  PubMed  Google Scholar 

  104. Duan ZX, Zhu PF, Dong H, Gu W, Yang C, Liu Q, et al. Functional significance of the TLR4/11367 polymorphism identified in Chinese Han population. Shock. 2007;28(2):160–4.

    CAS  PubMed  Google Scholar 

  105. Zhan X, Stamova B, Jin LW, DeCarli C, Phinney B, Sharp FR. Gram-negative bacterial molecules associate with Alzheimer disease pathology. Neurology. 2016;87(22):2324–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Reed-Geaghan EG, Savage JC, Hise AG, Landreth GE. CD14 and toll-like receptors 2 and 4 are required for fibrillar Abeta-stimulated microglial activation. J Neurosci. 2009;29(38):11982–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Reed-Geaghan EG, Reed QW, Cramer PE, Landreth GE. Deletion of CD14 attenuates Alzheimer’s disease pathology by influencing the brain’s inflammatory milieu. J Neurosci. 2010;30(46):15369–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Gale SC, Gao L, Mikacenic C, Coyle SM, Rafaels N, Murray Dudenkov T, et al. APOepsilon4 is associated with enhanced in vivo innate immune responses in human subjects. J Allergy Clin Immunol. 2014;134(1):127–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Mhatre-Winters I, Eid A, Han Y, Tieu K, Richardson JR. Sex and APOE genotype alter the basal and induced inflammatory states of primary microglia from APOE targeted replacement mice. Int J Mol Sci. 2022;23(17):9829.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Dela Justina V, Giachini FR, Sullivan JC, Webb RC. Toll-like receptors contribute to sex differences in blood pressure regulation. J Cardiovasc Pharmacol. 2020;76(3):255–66.

    CAS  PubMed  Google Scholar 

  111. Zhang C, Wang Y, Wang D, Zhang J, Zhang F. NSAID exposure and risk of Alzheimer’s disease: an updated meta-analysis from cohort studies. Front Aging Neurosci. 2018;10:83.

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  113. Przybyla GW, Szychowski KA, Gminski J. Paracetamol - an old drug with new mechanisms of action. Clin Exp Pharmacol Physiol. 2020.

  114. Sienski G, Narayan P, Bonner JM, Kory N, Boland S, Arczewska AA, et al. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci Transl Med. 2021;13(583):eaaz4564.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Wang N, Wang M, Jeevaratnam S, Rosenberg C, Ikezu TC, Shue F, et al. Opposing effects of apoE2 and apoE4 on microglial activation and lipid metabolism in response to demyelination. Mol Neurodegener. 2022;17(1):75.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Lee S, Devanney NA, Golden LR, Smith CT, Schwartz JL, Walsh AE, et al. APOE modulates microglial immunometabolism in response to age, amyloid pathology, and inflammatory challenge. Cell Rep. 2023;42(3):112196.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Kloske CM, Gearon MD, Weekman EM, Rogers C, Patel E, Bachstetter A, et al. Association between APOE genotype and microglial cell morphology. J Neuropathol Exp Neurol. 2023;82(7):620–30.

    CAS  PubMed  Google Scholar 

  118. Wang C, Lu J, Sha X, Qiu Y, Chen H, Yu Z. TRPV1 regulates ApoE4-disrupted intracellular lipid homeostasis and decreases synaptic phagocytosis by microglia. Exp Mol Med. 2023;55(2):347–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Lefevre N, Corazza F, Valsamis J, Delbaere A, De Maertelaer V, Duchateau J, et al. The number of X chromosomes influences inflammatory cytokine production following toll-like receptor stimulation. Front Immunol. 2019;10:1052.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Traish A, Bolanos J, Nair S, Saad F, Morgentaler A. Do androgens modulate the pathophysiological pathways of inflammation? Appraising the contemporary evidence. J Clin Med. 2018;7(12):549.

    PubMed  PubMed Central  Google Scholar 

  121. Romerio A, Peri F. Increasing the chemical variety of small-molecule-based TLR4 modulators: an overview. Front Immunol. 2020;11:1210.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Michaud JP, Halle M, Lampron A, Theriault P, Prefontaine P, Filali M, et al. Toll-like receptor 4 stimulation with the detoxified ligand monophosphoryl lipid A improves Alzheimer’s disease-related pathology. Proc Natl Acad Sci U S A. 2013;110(5):1941–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Cui W, Sun C, Ma Y, Wang S, Wang X, Zhang Y. Inhibition of TLR4 induces M2 microglial polarization and provides neuroprotection via the NLRP3 inflammasome in Alzheimer’s disease. Front Neurosci. 2020;14:444.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This manuscript is in memory of Dr. LaDu, who recently tragically passed away. She will be missed by all.

Funding

This study was funded by NIH (NIA) R21 AG044682-01. Additional funding to the LaDu lab included NIH-NIA grants (R01 AG058068, R01 AG057008), institutional funds from the College of Medicine at the University of Illinois, Chicago, and philanthropic support from Louis and Christine Friedrich. LMT is supported by NIH-NIA R01AG061114, R61NS114353 (LMT), and University of Illinois at Chicago Institutional funds.

Author information

Authors and Affiliations

Authors

Contributions

MJL and LMT designed the original study. FN (Innaxon Biosciences) provided the IAXO-101. JMY maintained the EFAD mouse colony, treated the mice and executed the behavioral tests. AV and DB supervised the biochemical and immunostaining experiments, respectively, including data analysis and figure design. AN assisted with performing experiments. MP analyzed the behavioral data. FP, FN and AV critically reviewed the manuscript, giving valuable feedback. DB, MJL and LMT wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Leon M. Tai.

Ethics declarations

Ethics approval and consent to participate

All experiments follow the University of Illinois at Chicago Animal Care and Use Committee protocols.

Consent for publication

Not applicable.

Competing interests

FN is the founder and owner of the privately held company Innaxon Biosciences. No other competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

Additional file 1: Supplementary figures.

Additional file 2:

Table 1. List of reagents and antibodies.

Additional file 3.

Raw data and statistical analysis results presented in the manuscript.

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

Balu, D., Valencia-Olvera, A.C., Nguyen, A. et al. A small-molecule TLR4 antagonist reduced neuroinflammation in female E4FAD mice. Alz Res Therapy 15, 181 (2023). https://doi.org/10.1186/s13195-023-01330-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13195-023-01330-6

Keywords