Abstract
BT75, a boron-containing retinoid, is a novel retinoic acid receptor (RAR)α agonist synthesized by our group. Previous studies indicated that activation of retinoic acid (RA) signaling may attenuate progression of Alzheimer’s disease (AD). Presently, we aimed to examine the anti-inflammatory effect of BT75 and explore the possible mechanism using cultured cells and an AD mouse model. Pretreatment with BT75 (1 μM–25 μM) suppressed the release of nitric oxide (NO) and IL-1β in the culture medium of mouse microglial SIM-A9 cells activated by LPS. BMS195614, an RARα antagonist, partially blocked the inhibition of NO production by BT75. Moreover, BT75 attenuated phospho-Akt and phospho-NF-κB p65 expression augmented by LPS. In addition, BT75 elevated arginase 1, IL-10, and CD206, and inhibited inducible nitric oxide synthase (iNOS) and IL-6 formation in LPS-treated SIM-A9 cells, suggesting the promotion of M1-to-M2 microglial phenotypic polarization. C57BL/6 mice were injected intracerebroventricularly (icv) with streptozotocin (STZ) (3 mg/kg) to provide an AD-like mouse model. BT75 (5 mg/kg) or the vehicle was intraperitoneally (ip) injected to icv-STZ mice once a day for 3 weeks. Immunohistochemical analyses indicated that GFAP-positive cells and rod or amoeboid-like Iba1-positive cells, which increased in the hippocampal fimbria of icv-STZ mice, were reduced by BT75 treatment. Western blot results showed that BT75 decreased levels of neuronal nitric oxide synthase (nNOS), GFAP, and phosphorylated Tau, and increased levels of synaptophysin in the hippocampus of icv-STZ mice. BT75 may attenuate neuroinflammation by affecting the Akt/NF-κB pathway and microglial M1-to-M2 polarization in LPS-stimulated SIM-A9 cells. BT75 also reduced AD-like pathology including glial activation in the icv-STZ mice. Thus, BT75 may be a promising anti-inflammatory and neuroprotective agent worthy of further AD studies.
Keywords: neurodegenerative disease, retinoic acid receptor, neuroinflammation, nitric oxide, microglial polarization, boron-containing retinoid
1. Introduction
Alzheimer’s disease (AD), the most common form of dementia, is a terminal, progressive disorder leading to memory loss, personality changes and the inability to communicate [1]. Currently, nearly 5.3 million Americans aged 65 and older have AD, 47 million people live with dementia globally, and it is estimated to increase more than threefold (~131 million) by 2050 [2]. Two well-recognized pathological hallmarks are extracellular amyloid-β (Aβ) plaques due to inappropriate digestion of amyloid precursor protein, and intracellular neurofibrillary tangles (NFT) due to hyperphosphorylated tau protein [3]. There is no effective cure for AD, and current treatments are only palliative. Owing to insidious onset, long incubation period, and various equivocal mechanisms, it is difficult to explore an effective AD therapy. Among various hypotheses regarding AD pathology, neuroinflammation has been one of the focuses of recent research [4–6].
Neuroinflammation is considered a pathophysiological process involved in various neurological and neurodegenerative diseases [7, 8]. Microglia activation provides the first line of defense against invading pathogens or injury-related products [9]. Under physiological conditions, microglia have branched morphology and high motility, which are beneficial for monitoring the microenvironment, pruning synapses, and timely removal of apoptotic neurons to maintain CNS homeostasis [10]. In the case of brain injury, microglia are the first to respond to this injury and become active [11]. Activated microglia are currently thought to differentiate into different phenotypes, including deleterious M1 and neuroprotective M2 phenotypes [12]. Specifically, “classically activated” M1 microglia typically release damaging pro-inflammatory cytokines such as IL-1β, TNF-α, and IL- 6, and reactive oxygen species and nitrogen (ROS and RNS) that exacerbate brain damage [13–15]. In contrast, the “alternatively activated” M2 phenotype secretes excess anti-inflammatory cytokines and trophic factors such as IL-4, IL-10, TGF-β, that resolve local inflammation and promote brain recovery [12]. Also, arginase 1 (Arg1), which converts arginine to polyamines, proline, and ornithine, can effectively outcompete iNOS, which uses the same substrate arginine, and downregulates production of pro-inflammatory mediator nitric oxide. Thus, iNOS and Arg1 represent a set of markers for M1 and M2 microglia [16]. CD206, also known as mannose receptor 1 or MR1, involved in the pinocytosis and the phagocytosis of immune cells, has been shown to be positively associated with M2 polarization and to play a role in regulating this phenotype [12,16]. In AD, while phagocytosis by microglia is known to play a major role in Aβ clearance, the presence of proinflammatory cytokines is known to block the phagocytic activity of microglia and may induce proinflammatory astrocytes [17–19]. These elevated inflammatory responses are associated with the cognitive decline and neuronal loss observed in AD patients and in the animal models [20, 21]. Therefore, inhibiting M1 activation and promoting M2 activation is a promising strategy for the treatment of neuroinflammation-related diseases such as AD and ischemic stroke compared with general inhibition of microglial activation.
Retinoic acid receptors (RARs), the nuclear receptor superfamily, are involved in the regulation of cell differentiation, proliferation, embryonic development, metabolism, and other life activities [22]. Activation of the retinoic acid (RA) signaling pathway may attenuate progression of AD through up-regulating non-amyloidogenic α-secretase and down-regulating amyloidogenic β-secretase 1 [23]. One study showed RA, the main active vitamin A metabolite, acted primarily through CDK5 to inhibit APP processing and reduce tau phosphorylation [24]. RA is also implicated in exerting anti-inflammatory functions [25]. A recent study indicating the relationship between reduction in BACE1 expression and anti-inflammatory effects of RAR agonists underscores the importance of elucidation of mechanisms of anti-inflammatory effects of RAR agonists [26].
BT75, a boron-containing retinoic acid receptor α (RARα) agonist, was previously synthesized by Dr. Das as a derivative of an RARα agonist Am580 and has been shown to be less toxic compared to all-trans retinoic acid (ATRA) or Am580 [27]. The use of boron atoms in pharmaceutical drug design has a high potential for discovery of new biological activities [28]. In the current studies, we examined possible anti-inflammatory action of BT75 and its mechanisms, using microglial cell culture and an icv-STZ AD mouse model.
2. Materials and Methods
2.1. Synthesis of BT75
Synthesis of BT75 [Fig. 1; (E)-2-(4-(2-(6,8-dichloro-2-phenyl-2H-chromen-3-yl)vinyl)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane] was performed as follows: A clean oven dried three-neck round bottom flask (RBF) was charged with aldehyde 1 [27] (1 equiv.) in DMF. Then addition of salt 2 (1.2 equiv.) Sodium tertbutoxide (STB) (3 equiv.) was added into the reaction mixture at 0°C. The reaction mixture was stirred at room temperature for 24 h. Progress of the reaction was monitored by TLC (100% Hexane), In TLC nonpolar spot was observed corresponding to aldehyde. After completion of the reaction, reaction mixture was neutralized by 2N HCl. After neutralization, ethyl acetate was added in the reaction mixture; the combined organic layer was collected and dried with Na2SO4 and evaporated under reduced pressure. The crude material was purified by column chromatography (5% ethyl acetate : 95 % hexane). After purification white solid was observed (BT75). The result of BT synthesis was: ( Yield = 72 %), 1H NMR (600 MHz, DMSO-d6) δ 7.61 (d, J = 7.8 Hz, 2H), 7.47 (d, J = 7.8 Hz, 2H), 7.42 (d, J = 7.2 Hz, 2H), 7.35 – 7.29 (m, 5H), 7.23 (d, J = 16.4 Hz, 1H), 7.01 (s, 1H), 6.71 (d, J = 16.5 Hz, 1H), 6.64 (s, 1H), 1.26 (s, 12H).
Fig. 1.

BT75 chemical structure and its synthetic formula.
2.2. Other chemicals
BMS195614, RARα antagonist, was purchased from Tocris (Minneapolis, MN). LPS (0111:B4, Cat# LPS25) and streptozotocin (Cat# S0130) were purchased from Sigma (St. Lous, MO). The 3-(4,5-dimethyl-2-thiazolyl) 2,5-diphenyl-2H-tetrazoliumbromide (MTT) was obtained from Sigma [Roche Cell Proliferation Kit I (MTT)].
2.3. Cell culture
Mouse microglial cell line SIM-A9 was obtained from ATCC (CRL3265, Manassas, VA) and cultured in DMEM/F12 containing 10% fetal bovine serum, 5% horse serum and 1% penicillin-streptomycin solution. Neuro-2a cell line was also purchased from ATCC (CCL-131) and cultured in DMEM containing 10% fetal bovine serum and 1% penicillin-streptomycin solution. All cells were cultured under the conditions of 37°C and 5% CO2 in an incubator.
BT75 (from 25 μM to 1 μM) used for the treatment of SEM-A9 cells was dissolved in DMSO followed by dilution in DMEM. Final concentrations of DMSO were less than 0.1%. Other reagents (LPS and STZ) used were dissolved in DMEM.
2.4. AD-like icv-STZ model
Three-month-old male C57BL/6 mice were purchased from Jackson Laboratory (Bar Harbor, ME). All procedures described below were approved by the Nathan Kline Institute IACUC and were in accordance with NIH guidelines for the proper treatment of animals. The mice were injected (icv) with STZ at 3 mg/kg body weight to induce AD-like model using a slightly modified version of previously reported methods [29–31]. The scalp of the mouse was incised to locate bregma. Small burr holes (1 mm in diameter) were drilled, and STZ in citrate buffer (50 mM, pH 4.5) (volume was calculated according to the mouse weight) was injected into both sides of the lateral ventricle with a microsyringe fitted with a 26-gauge blunt cannula at a rate of 0.5 μL/min with the following coordinates: 0.2 mm posterior to the bregma, ±1.0 mm lateral to the sagittal line, and 2.5 mm ventral to the surface of the skull. The cannula was withdrawn after remaining for 5 min in the injection position. The incision was closed with wound clips, and the animal was allowed to recover under a heat lamp. Sham mice had received the same surgical procedures except with injection of the same volume of the vehicle (50 mM citrate buffer).
Three days after the icv-STZ injection, BT75 (in 5% DMSO in saline, 5mg/kg body weight, STZ+BT75 group) or the vehicle (5% DMSO in saline) (STZ group) was given to the mice with intraperitoneal (ip) injection once a day for 3 weeks. Sham mice were also given vehicle (Control group) or BT75 (5 mg/kg body weight, BT75 group). After drug administration, the hippocampus was collected for western blot or the brain was perfusion-fixed for immunofluorescence staining.
2.5. Cell viability detection
MTT method was used for cell viability detection. SIM-A9 cells (2 × 105 cells/well) were propagated in 96-well plates. SIM-A9 cells were treated with different doses of BT75 for 24 h or pretreated with different doses of BT75 (25, 10, 5 and 1μM) for 30 min and then subjected to 24 h incubation with 1 μg/ml LPS. MTT was added to cell culture with the final concentration of 0.5 mg/ml. After 4 h incubation, the cell media was removed and 100 μl DMSO was added to dissolve the crystal. The absorbance was measured at 570 nm with reference wave at 630 nm. Neuro-2a cells were treated with BT75 (1 mM, 100 μM, 10 μM and 1 μM) or STZ (1 mM, 500 μM, 100 μM, 50 μM and 10 μM) for 24 h, and the cell viability was detected by MTT. The cell viability of Neuro-2a cells pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with STZ (1 mM) for another 24 h. Cell viability was assessed using the MTT assay.
2.6. Nitric Oxide (NO) detection
Griess reaction method was used for the detection of NO production. Due to its extreme instability, NO is quickly and proportionally metabolized to nitrite. Therefore, nitrite has been extensively used as an indicator of NO. SIM-A9 cells (2 × 105 per well) were cultured in 96-well plates overnight. Then the cells were subjected to 30 min incubation of BT75 (25, 10, 5 and 1 μM) followed by 24 h stimulation with 100 ng/mL LPS. Same volume of the cell culture medium and Griess agent solution (Sigma-Aldrich, Cat# G4410) were mixed together and the absorbance at 540 nm was detected after 30 min.
2.7. Inflammatory cytokine detection
Enzyme-Linked Immunosorbent Assay (ELISA) was used to detect inflammatory factors in the cell culture medium. SIM-A9 cells (2 × 105 per well) were cultured in 96-well plates overnight. Then the cells were pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with LPS (100ng/mL) for another 24 h. TNF-α, IL-1β l, and IL-10 levels in the cell culture medium were detected by commercial ELISA kits, TNFα Quantikine ELISA kit (R&D Systems, Minneapolis, MN, Cat# MTA00B ), mouse IL-10 Quantikine ELISA kit (R&D Systems), and IL-1β mouse ELISA kit (Thermo Fisher Scientific, Watham, MA, Cat# BMS6002), respectively, according to the manufacturers’ instructions.
2.8. Western blot
Western blot assay was done for the detection of target proteins involved in the cell signal pathway both in SIM-A9 cells and brain hippocampus tissue. Cells were pretreated with BT75 (10μM) for 30 min before 100 ng LPS treatment for 24 h, then the cells were collected and lysed with RIPA lysis buffer plus protease and phosphatase inhibitors to detect Akt, phospho (p)-Akt, NF-κB p65, p-NF-κB p65, iNOS, Arginase 1 (Arg1), IL-6, CD206, and β-actin expression by western blot. The hippocampus was also lysed and nNOS, GFAP, synaptophysin, p-Tau and β-actin expression were detected by western blot. Briefly, total protein amounts were measured by the Pierce BCA protein assay, and 20 μg amount of total protein from each sample was separated by SDS-PAGE gel and transferred to nylon membranes. After blocking with 5% non-fat milk powder in TBST for 2 h at room temperature, these membranes were incubated with primary antibodies against Akt (Cat#4685), p-Akt (Cat#4060), NF-κB p65 (Cat#8242), p-NF-κB p65 (Cat#8242), iNOS (Cat#13120), Arg1 (Cat#93668), β-actin (Cat#3700), nNOS (Cat#4231), GFAP (Cat#3670), p-Tau (p-Thr231, Cat#71429), synaptophysin (Cat#36406), CD206/MRC1 (Ca#24595), and IL-6 (#12912) [all from Cell Signaling Technology (Danvers, MA) with dilution of 1:1000] and corresponding secondary antibodies [Millipore Sigma (Burlington, MA) with dilution of 1:2500]. The bands were shown with an enhanced chemiluminescence (ECL) method and analyzed by Image J software.
2.9. Immunofluorescence staining
Immunofluorescence staining was done as previously described [32]. Mice were perfused with a solution containing 4% paraformaldehyde and 4% sucrose in cacodylate buffer (pH 7.2), and the heads were removed and further fixed in the perfusion solution overnight. Then brains were removed, transferred to phosphate buffered saline (PBS) solution, and kept at 4°C for 2–5 days until cut with a vibratome into 50 μm thick coronal sections. The free-floating sections were rinsed in PBS, permeabilized in methanol for 10 min, and incubated for 30 min in blocking solution (PBS containing 5% BSA and 0.1% Triton X-100), followed by incubation overnight with antibodies against GFAP (GA5) (mouse mAb Cat#3670, Cell Signaling with dilution of 1:500) and Iba1 (rabbit polyclonal Cat#019–19741, FujifilmWako Chemicals, Richmond, VA with dilution of 1:500), rinsed in PBS three times, followed by incubation with Alexa Fluoro594 goat anti-rabbit IgG and Alexa Fluoro488 goat anti-mouse IgG (Life Technologies, Grand Island, NY) in 0.1% Triton X-100 in PBS containing 1% BSA for 1 h at r.t.. Sections were finally rinsed in PBS three times, mounted, and coverslipped using ProLong Gold Antifade Reagent (Life Technologies). Glial activation was evaluated in the dorsal hippocampus region, especially in the fimbria, where activation was prominent. For quantitative measurement, GFAP-positive cell number or rod (or amoeboid)-like Iba1-positive cell number was counted in the hippocampal fimbria using 3 sections (around bregma −1.34 to −1.82) from each brain, and 3–4 brains per each treatment group were analyzed. Cell counting and the measurement of area of interest (AOI) were done by Image J software.
2.10. Statistical analysis
Data were presented in the form of mean ± SD. One-way analysis of variance (ANOVA) with a Tukey test was performed to analyze the data using GraphPad Prism software, and p < 0.05 was defined as statistically significant.
3. Results
3.1. Effects of BT75 on cell viability of SIM-A9 cells treated by a high dose of LPS
The cytotoxicity test of BT75 (500, 250, 100 and 50 μM) was done using SIM-A9 cells. BT75 was dissolved in DMSO followed by dilution in DMEM. Final concentrations of DMSO were less than 0.1%. the results showed that 50 μM BT75 was not toxic to cells (Fig. 2A). Therefore, we chose lower than 50 μM BT75 for further experiments. The cell viability of SIM-A9 cells pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with LPS (1 μg/mL) for another 24 h indicated that BT75 partially inhibited the decrease in cell viability induced by high dose of LPS (Fig. 2B).
Fig. 2.

Effects of BT75 on the cytotoxicity and cell viability of SIM-A9 cells stimulated by LPS. (A) Cell viability of SIM-A9 cells treated with BT75 (500, 250, 100 and 50 μM) for 24 h. The results are expressed as the mean ± SD (n = 8). **p < 0.01 as compared with control group. (B) The cell viability of SIM-A9 cells pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with LPS (1 μg/mL) for another 24 h. Cell viability was assessed using the MTT assay. The results are expressed as the mean ± SD (n = 8). ##p < 0.01 as compared with control group, **p < 0.01 as compared with LPS-treated group.
3.2. Effects of BT75 on NO production and inflammatory factors induced by LPS in SIM-A9 cells
SIM-A9 cells were pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with LPS (100 ng/mL) for another 24 h. LPS treatment increased NO production in the cell culture medium, and BT75 inhibited NO production at a dose-dependent manner (Fig. 3A). TNF-α and IL-1β levels detected by ELISA in the cell culture medium also increased in the LPS group, and BT75 at the concentrations of 25 and 10 μM was effective in decreasing IL-1β levels induced by LPS (Fig. 3C), but not TNF-α levels at these concentrations (Fig. 3B). In contrast, IL-10 levels in the cell culture medium did not change by LPS, but significantly increased by BT75 treatment (Fig. 3D).
Fig. 3.

Effects of BT75 on NO production and inflammatory factors induced by LPS in SIM-A9 cells. SIM-A9 cells were pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with LPS (100 ng/mL) for another 24 h. (A) NO production in the cell culture medium was assessed by the Griess method. TNF-α (B), IL-1β (C), and IL-10 (D) levels in the cell culture medium were detected by ELISA. The results are expressed as the mean ± SD (n = 4–5). ##p < 0.01 as compared with control group, *p < 0.05 and **p<0.01 as compared with LPS-treated group.
3.3. BT75 may ameliorate inflammation induced by LPS in SIM-A9 cells via Akt/NF-κB pathway
SIM-A9 cells pretreated with BT75 (10 μM) or the vehicle for 30 min were cultured in the presence or absence of 100 ng/ml LPS for 24 h. Then, the expression of p-Akt, Akt, p-NF-κB p65, NF-κB p65 and β-actin in the cells were examined by Western blot. Fig. 4 showed the ratio of p-Akt/Akt and p-NF-κB p65/NF-κB p65 increased in the LPS group as compared with control group, and BT75 prevented the increase.
Fig. 4.

BT75 may ameliorate inflammation induced by LPS in SIM-A9 cells via regulation of the AKT/NF-κB pathway. Cells were pretreated with BT75 (10μM) for 30 min before 100 ng/ml LPS treatment for 24 h. Akt/p-AKT and NF-κb/p- NF-κb expression in the cells were detected by Western blot. (A) Represented immunoblot bands for p-Akt, Akt, p- NF-κB, NF-κB, and β-actin. (B and C) Quantitative analysis of immunoblot bands. Protein expression levels were normalized to β-actin. The data were expressed as mean ± SD (n = 3–4). ##p < 0.01 as compared with control group, *p < 0.05 as compared with LPS-treated group.
3.4. BT75 may ameliorate inflammation induced by LPS in SIM-A9 cells via promoting M1 to M2 phenotypic polarization
SIM-A9 cells pretreated with BT75 (10 μM) or the vehicle for 30 min were further incubated with 100 ng LPS for 24 h, and iNOS, IL-6, Arg1, and CD206 expression in the cells were analyzed by Western blot. The results showed that LPS treatment increased the expression of iNOS and IL-6, while BT75 inhibited iNOS and IL-6 expression (Fig. 5A, B and C). Interestingly, BT75 alone totally inhibited iNOS expression (Fig. 5A and B). At the same time, we found that Arg1 and CD206 expression decreased in the LPS group, and BT75 inhibited this reduction (Fig. 5A, D and E). iNOS and IL-6 are considered M1 markers, and Arg1 and CD206 are considered M2 markers. These results, combined with the results of ELISA for IL-1β and IL-10 expression (Fig. 3), suggest that BT75 promoted microglia from M1 to M2 phenotypic polarization.
Fig. 5.

BT75 ameliorated inflammation induced by LPS in SIM-A9 cells via promoting M1-M2 phenotypic polarization. Cells were pretreated with BT75 (10 μM) for 30 min before 100 ng/ml LPS treatment for 24 h. iNOS, IL-6, Arg1, and CD206 expression in the cells were detected by Western blot. (A) Representative immunoblots for iNOS, IL-6, Arg-1, CD206, and β-actin. (B to E) Quantitative analysis of immunoblot bands. Protein expression levels were normalized to β-actin. The data were expressed as mean ± SD (n = 3–4). ##p < 0.01 as compared with control group, **p < 0.01 as compared with LPS-treated group.
3.5. BMS195614, a selective RARα antagonist, partially blocked the inhibition of NO production by BT75 in SIM-A9 cells activated by LPS
BMS195614 (10 μM and 20 μM) was administrated for 24 h and MTT assay showed that 20 μM BMS195614 had a little toxic effect on cells (Fig. 6A). The cells were pretreated with BMS195614 (10 μM and 20 μM) for 30 min, then treated with 100 ng LPS for another 24 h, and NO production in the cell medium was measured. The results showed that BMS195614 had no effect on NO production in LPS-stimulated SIM-A9 cells (Fig. 6B). In Fig. 6C, after pretreatment with BMS195614 (10 μM) for 30 min, cells were incubated in the presence of BT75 (10μM) or the vehicle for 30 min, and further treated with 100 ng LPS for another 24 h. The result indicates that BMS195614 incompletely but significantly blocked the inhibition of NO production by BT75 in LPS-stimulated SIM-A9 cells, suggesting that BT75 inhibited LPS-induced NO production at least partially through the RARα activation.
Fig. 6.

BMS195614, an RARα antagonist, partially blocked the inhibition of NO production by BT75 in LPS-activated SIM-A9 cells. Cells were pretreated with BMS195614 (10 μM) for 30 min and then incubated with BT75 (10 μM) for another 30 min. Then, LPS (100 ng/ml) was added and incubated for 24 h. (A) The cytotoxicity test of BMS195614. (B) Effect of BMS195614 on NO production in LPS-induced SIM-A9 cells. (C) BMS195614 partially blocked the inhibition of NO production by BT75 in LPS-activated SIM-A9 cells. The data were expressed as mean ± SD (n = 4). ##p < 0.01 as compared with control group, **p < 0.01 as compared with LPS-treated group. &&p<0.01 as compared to LPS+BT75 group.
3.6. BT75 inhibited icv-STZ-induced glial activation in mouse brain hippocampus
Double Immunofluorescence staining on mouse brain sections was performed for the detection of GFAP-positive astrocytes and Iba1-positive microglia in the dorsal hippocampus area. In Fig. 7A, brighter staining by GFAP was shown in the hippocampus in STZ group compared to control group, while BT75 inhibited this increase. BT75 alone was not different from the control group. Fig. 7A also showed that STZ increased Iba-1 positive microglia especially in the fimbria of hippocampus, while BT75 treatment decreased STZ-stimulated Iba-1 positive microglia. As compared to control group, BT75 alone had no effect on Iba-1 positive microglia. In the fimbria, STZ increased rod or amoeboid-like Iba1-positive cells with large cell body, and those seemingly activated microglia were reduced by BT75 treatment (Fig. 7B). The quantitative measurement of the densities of the activated cells in the fimbria (Fig. 7C) indicates that BT75 significantly reduced the activated cell densities. We also measured densities of GFAP+ astrocytes in the fimbria (Fig. 7D), which indicate that BT75 reduced GFAP+ cell densities augmented by STZ treatment.
Fig. 7.

BT75 reduced icv-STZ-induced glial activation in mouse brain. (A) GFAP-positive astrocytes and Iba1-positive microglia in the dorsal hippocampus are shown here. The representative image from each group, Ctr (n=3), STZ (n=3), STZ+BT75 (n=3), and BT75 (n=4) group, is shown here. The bar indicates 500 μm. (B) Iba1-positive microglia in the hippocampus fimbria are shown. The bar indicates 50 μm. (C) Larger round or rod-like cell densities in the fimbria were compared among the control, STZ, STZ+BT75, and BT75 groups. * indicates that the STZ group is different from all other groups significantly (p<0.05). (D) The densities of GFAP-positive cells were compared among the control, STZ, STZ+BT75, and BT75 groups. * indicates that the STZ group is different from all other groups significantly (p<0.05).
3.7. Effect of BT75 on nNOS, GFAP, Synaptophysin and p-Tau expression in the hippocampus of icv-STZ-treated mice
Western blot was done for the analyses of GFAP, synaptophysin, nNOS and p-Tau. The results indicated that BT75 inhibited glial cell activation and p-Tau expression in the hippocampus of icv-STZ mice. BT75 also increased synaptophysin and decreased nNOS expression in the hippocampus of icv-STZ mice (Fig. 8A–E).
Fig. 8.

Effect of BT75 on nNOS, GFAP, synaptophysin and p-Tau expression in the hippocampus of icv-STZ-treated mice. (A) Representative immunoblot bands for synaptophysin and p-Tau and β-actin. (B and C) Quantitative analysis of immunoblot bands. Protein expression levels were normalized to β-actin. The data were expressed as mean ± SD (n = 3). #p < 0.05 as compared with control group, *p < 0.01 as compared with STZ-treated group.
3.8. BT75 had no protective effect on STZ-induced Neuro-2a cytotoxicity
Neuro-2a cells were treated with BT75 (1mM, 100μM, 10μM and 1 μM) for 24 h and cell viability was detected. The results showed that the concentration of BT75 lower than 100 μM was not toxic to Neuro-2a cells (Fig. 9A). STZ (1mM, 500μM, 100μM, 50μM and 10 μM) was administrated to Neuro-2a cells for 24 h to see the cell damage. In Fig. 9B, the cell viability decreased at a dose dependent manner, and 1mM STZ inhibited cell viability to 74% of the control. Neuro-2a cells were pretreated with BT75 (25, 10, 5 and 1 μM) for 30 min followed by treatment with STZ (1 mM) for another 24 h. The cell viability was determined by MTT. The results showed BT75 had no effect on the decreased cell viability induced by STZ, indicating that BT75 may exert the protective effect not directly through neurons injured by STZ (Fig. 9C).
Fig. 9.

BT75 had no protective effect on STZ-induced Neuro-2a cytotoxicity. (A) Cell viability of Neuro-2a cells treated with BT75 (1mM, 100μm, 10μm and 1 μM) for 24 h. (B) Cell viability of Neuro-2a cells treated with STZ (1mM, 500μm, 100μm, 50μm and 10 μM) for 24 h. (C) The cell viability of Neuro-2a cells pretreated with BT75 (25, 10, 5 and 1 μM) for half an hour followed by treatment with STZ (1 mM) for another 24 h. Cell viability was assessed using the MTT assay. The results are expressed as the mean ± SD (n = 4). **p < 0.01 as compared with control group.
4. Discussion
Several retinoid-based drugs such as tamibarotene, bexarotene and acitretin, which activate RA signaling, have been repurposed for treatments of CNS diseases like AD [33–35]. These drugs have shown promising effects on improvement of AD pathology in mainly animal models by reducing Aβ production and neuroinflammation. To improve the efficacy and safety of those retinoid-based compounds, we synthesized BT75, a boron-containing RARα agonist. BT75, a derivative of the RARα-specific agonist Am580, binds to RARα with a higher affinity and exhibits less toxicity than Am580 and all-trans RA (ATRA) [28]. The use of boron atoms in pharmaceutical drug design has a high potential for discovery of new biological activities. Boron-containing compounds have been shown to have antioxidant and neuroprotective properties in nature by scavenging Reactive Oxygen Species (ROS) to produce carbon-oxygen bond [36]. Coban et. al showed that boron decreases malathion-induced oxidative stress, enhances the antioxidant defense mechanism and regenerates tissues in rats [37]. In this study, we aimed to explore the anti-inflammation effect and its mechanisms of BT75, and its treatment possibility for AD, since recent studies suggest that anti-inflammatory effects of RARα agonists may attenuate AD pathology [38].
We examined anti-inflammatory effects of BT75 using mouse microglial cell line, SIM-A9, because this spontaneously immortalized cell line stably exhibits microglial phenotypes, including expression of Iba1 and CD68, secretion of inflammatory mediators, and phagocytic activity, which are similar to cultured primary microglia [39, 40]. We found that BT75 attenuated NO production, elevation in the iNOS level, and proinflammatory cytokine IL-1β secretion induced by LPS in SIM-A9 cells (Figs. 3 and 5). This agrees with previous studies indicating that retinoic acid (RA) exerts potent anti-inflammatory effects under conditions of neuroinflammation. RA levels decrease in LPS-activated primary cultured microglia by increased protein expression of RA degradative enzymes, and the pharmacological inhibition of intracellular RA degradation or addition of ATRA exerts potent anti-inflammatory effects in these microglia [41, 42]. Also, inflammatory chemokine release of cultured astrocytes treated with LPS is reduced by ATRA [43]. RARα activation significantly decreased TNFα release from Aβ-treated cultured microglia and induces Aβ clearance in these cultures [44]. In AD mouse models, ATRA has been shown to reduce microglial and astrocytic activation, although these effects may be due to reduced Aβ levels by ATRA [44].
Our study also showed that BT75 decreased p-AKT and p-NF-κB p65 expression enhanced by LPS in these cells (Fig. 4). NF-κB is a family of nuclear transcription factors that play a dominant role in the pathogenesis of many chronic inflammatory processes and regulation of immune responses and is involved in the pro-inflammatory activation of microglia and astrocytes [45]. Normally, NF-κB is in the cytoplasm, and once activated, phosphorylated NF-κB dimers are released and translocated to the nucleus, where they induce cytokines, iNOS, and NO production [46]. It has been reported that the levels of NF-κB p65 are significantly elevated in the brains of AD patients [47], and NF-κB activity also increases in AD mouse models [48, 49]. Therefore, inhibition of NF-κB activation in microglia can modulate the progression of AD caused by neuroinflammation. Our results indicate that BT75 inhibited the activation of the NF-κB pathway, thereby inhibiting LPS-induced neuroinflammation in SIM-A9 cells. We also observed that BT75 induced a small but significant decrease in p-Akt augmented by LPS treatment (Fig. 4). Since Akt is shown to be the upstream signal molecule of NF-κB/IκBα [50], BT75 may ameliorate inflammation induced by LPS in SIM-A9 cells via the Akt/NF-κB pathway.
Our results also suggest that BT75 promoted M1-to-M2 phenotypic polarization in LPS-stimulated SIM-A9 cells. Microglia are classified as M1/M2 after activation, which helps to understand the functional status of microglia during the progression of injury and helps us explore new therapeutic strategies, although there are limitations of the M1/M2 classification system because of much higher heterogeneity in functions and morphologies found in microglia [51]. Previous literature supports the dual role of polarized microglial populations in a variety of neurological diseases, such as focal stroke and AD [52, 53]. Since activation of microglia with an anti-inflammatory M2 phenotype leads to brain repair and regeneration, inhibition of M1 activation and promotion of M2 activation is a promising strategy for the treatment of neuroinflammation-related diseases compared to inhibition of microglia activation in general. The present study showed that BT75 increased an M2 marker, Arg1, and decreased M1 marker, iNOS, in SIM-A9 cells activated with LPS (Fig. 5). Since Arg1 and iNOS utilize the same substrate arginine, and Arg1 can effectively outcompete iNOS to reduce NO production, iNOS and Arg1 are considered a typical set of M1 and M2 markers. In addition, our studies indicated that proinflammatory factors, IL-1 and IL-6, elevated by LPS were reduced by BT75 treatment, and levels of IL-10 and CD206, which are considered M2 markers, increased by BT75. These results suggest that BT75 may exert anti-inflammatory activity by promoting M1-to-M2 phenotypic polarization.
BMS195614, a specific RARα antagonist, partially blocked the inhibition of NO production by BT75 in LPS-induced SIM-A9 cells (Fig. 6), indicating that BT75 inhibited LPS-induced NO production at least partially through RARα activation. The cause of the incomplete action of BMS195614 is presently unknown. Experimental conditions such as concentrations of antagonists and length of the preincubation time may influence the result. Furthermore, since RA is indicated to elicit non-canonical (RAR/RXR-independent) activities beyond regulating gene expression [54], the possibility of such RAR/RXR-independent activity of BT75 cannot be eliminated.
In our in vivo experiment, we used 3 month-old male mice injected via icv with STZ (3 mg/kg) once. Although STZ has been originally used peripherally to induce animal models of diabetes, STZ injection via icv at a low dosage to rodents does not alter peripheral glucose levels and is used as an experimental model for sporadic AD. These mice show AD-like features including disturbances in brain insulin signaling and glucose utilization, accumulation of Aβ plaques, tau phosphorylation, inflammation, and progressive cognitive impairments [55–57]. Compared to transgenic mouse models, the icv-STZ model shows more enhanced neuroinflammation, and the effects of anti-inflammatory agents have been tested using the icv-STZ model [58,59]. Therefore, we examined the effects of BT75 in this icv-STZ mouse model.
Immunofluorescence staining showed that BT75 administration inhibited icv-STZ-induced activation of microglia (Iba-1) and astrocytes (GFAP) mainly in the hippocampus (Fig. 7). Microglia are resident macrophages widely distributed in the brain, and Iba-1 is a marker of microglial activation. Reactive astrocytes are characterized by increased expression of glial fibrillary acidic protein (GFAP). Activated microglia and astrocytes are known to be involved in Aβ deposition and neurofibrillary tangle accumulation during AD progression [60, 61]. Western blot experiments also showed that BT75 decreased levels of GFAP enhanced in hippocampus of icv-STZ mice (Fig. 8).
Tau is the major microtubule-associated protein (MAP) of normal mature neurons and is essential for the assembly and stabilization of the structural integrity of microtubules [62]. In the brains of AD patients, tau is more hyperphosphorylated than normal neurons, loses its ability to bind to microtubules, and subsequently aggregates into pairs of helical filaments that mix with straight filaments to form neurofibrillary tangles [63]. Our results showed that BT75 attenuated p-Tau expression in the hippocampus of icv-STZ mice. BT75 also increased synaptophysin suggesting the better synaptic integrity by BT75 and decreased nNOS expression in the hippocampus of icv-STZ mice. Thus, BT75 showed anti-inflammatory activity not only in vitro but also in vivo and reduced some of the AD-like neuropathology observed in icv-STZ mice. However, we may need to increase sample size of the animal experiments to evaluate possible M1-to-M2 phenotypic polarization of microglia by BT75 in vivo. Also, further studies which include female samples, would be necessary because sex differences in immune responses, including sex differences in microglial functionality, have been reported [64].
It has been shown that in vitro STZ decreases cell viability and choline levels, and increases acetylcholinesterase activity, tau phosphorylation and amyloid aggregation in N2a cells, and is used as a model for analyzing diabetes or AD-related cellular processes [65, 66]. We found that BT75 had no significant protective effect against STZ-induced Neuro-2a cytotoxicity. These results indicate that BT75 may not exert its direct neuroprotective effect against STZ-induced toxicity in neurons.
These results, together with the literature indicating that RARα agonists attenuate AD pathology suggest that BT75 would be a promising small molecule for treatment of diseases caused or exacerbated by neuroinflammation [26, 33, 44, 67].
5. Conclusion
BT75, a novel RARα agonist, attenuated the cytotoxicity induced by LPS in SIM-A9 cells and suppressed the releases of NO and IL-1β induced by LPS in SIM-A9 cells. The anti-inflammatory effects of BT75 may be mediated by the AKT/NF-κB pathway. Also, BT75 may promote microglial M1 to M2 polarization, because BT75 elevated Arg1, IL-10, and CD206, and inhibited iNOS and IL-6 expression in LPS-treated SIM-A9 cells. Also, BT75 shows anti-inflammatory effects in the icv-STZ-induced AD mouse model. BT75 reduced densities of GFAP-positive astrocytes and rod or amoeboid-like Iba1-positive cells, which were elevated in the hippocampal fimbria of icv-STZ mice. Furthermore, BT75 ameliorated AD-like pathology by decreasing tau phosphorylation and increasing synaptophysin expression in the icv-STZ mouse hippocampus. Thus, BT75 is a promising anti-inflammatory agent worthy of further studies towards treatment of neuroinflammation-related diseases, such as AD.
Acknowledgement
This work was supported by grants from National Institute on Alcohol Abuse and Alcoholism (R21AA027374 to B.C.D and M.S; R01AA023181 to M.S., D.A.W., and J.F.S).
Funding:
This work was supported by grants from National Institute on Alcohol Abuse and Alcoholism (R21AA027374 to B.C.D and M.S; R01AA023181 to M.S., D.A.W., and J.F.S).
Footnotes
Competing Interests: The authors have no relevant financial and non-financial interests to disclose.
Ethics approval: All procedures for animal studies were approved by the Nathan Kline Institute IACUC and were in accordance with NIH guidelines for the proper treatment of animals.
Data availability:
All data and materials are available in the article or upon request to corresponding authors.
References
- 1.Jessen F, Amariglio RE, Buckley RF, van der Flier WM, Han Y, Molinuevo JL, Rabin L, Rentz DM, Rodriguez-Gomez O, Saykin AJ et al. : The characterisation of subjective cognitive decline. Lancet Neurol 2020, 19(3):271–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M: Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine 2019, 14:5541–5554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lashley T, Schott JM, Weston P, Murray CE, Wellington H, Keshavan A, Foti SC, Foiani M, Toombs J, Rohrer JD et al. : Molecular biomarkers of Alzheimer’s disease: progress and prospects. Dis Model Mech 2018, 11(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Revi M: Alzheimer’s Disease Therapeutic Approaches. Adv Exp Med Biol 2020, 1195:105–116. [DOI] [PubMed] [Google Scholar]
- 5.Ozben T, Ozben S: Neuro-inflammation and anti-inflammatory treatment options for Alzheimer’s disease. Clin Biochem 2019, 72:87–89. [DOI] [PubMed] [Google Scholar]
- 6.Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R: Inflammation: the link between comorbidities, genetics, and Alzheimer’s disease. J Neuroinflammation 2018, 15(1):276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Heppner FL, Ransohoff RM, Becher B: Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci 2015, 16(6):358–372. [DOI] [PubMed] [Google Scholar]
- 8.Spangenberg EE, Green KN: Inflammation in Alzheimer’s disease: Lessons learned from microglia-depletion models. Brain Behav Immun 2017, 61:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cai Z, Hussain MD, Yan LJ: Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int J Neurosci 2014, 124(5):307–321. [DOI] [PubMed] [Google Scholar]
- 10.Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC: Neuronal Cell Death. Physiol Rev 2018, 98(2):813–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Li Q, Wu Y, Chen J, Xuan A, Wang X: Microglia and immunotherapy in Alzheimer’s disease. Acta Neurol Scand 2022, 145(3):273–278. [DOI] [PubMed] [Google Scholar]
- 12.Sanjay Shin JH, Park M, Lee HJ: Cyanidin-3-O-Glucoside Regulates the M1/M2 Polarization of Microglia via PPARgamma and Abeta42 Phagocytosis Through TREM2 in an Alzheimer’s Disease Model. Mol Neurobiol 2022, 59(8):5135–5148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang F, Zhong R, Li S, Fu Z, Cheng C, Cai H, Le W: Acute Hypoxia Induced an Imbalanced M1/M2 Activation of Microglia through NF-kappaB Signaling in Alzheimer’s Disease Mice and Wild-Type Littermates. Front Aging Neurosci 2017, 9:282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kirkley KS, Popichak KA, Afzali MF, Legare ME, Tjalkens RB: Microglia amplify inflammatory activation of astrocytes in manganese neurotoxicity. J Neuroinflammation 2017, 14(1):99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Li L, Li L, Zhang J, Huang S, Liu W, Wang Z, Liang S, Tao J, Chen L: Disease Stage-Associated Alterations in Learning and Memory through the Electroacupuncture Modulation of the Cortical Microglial M1/M2 Polarization in Mice with Alzheimer’s Disease. Neural Plast 2020, 2020:8836173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation 2014,11:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tuppo EE, Arias HR: The role of inflammation in Alzheimer’s disease. Int J Biochem Cell Biol 2005, 37(2):289–305. [DOI] [PubMed] [Google Scholar]
- 18.Weitz TM, Town T: Microglia in Alzheimer’s Disease: It’s All About Context. Int J Alzheimers Dis 2012, 2012:314185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC et al. : Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541(7638):481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cho SH, Sun B, Zhou Y, Kauppinen TM, Halabisky B, Wes P, Ransohoff RM, Gan L: CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease. J Biol Chem 2011, 286(37):32713–32722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cabinio M, Saresella M, Piancone F, LaRosa F, Marventano I, Guerini FR, Nemni R, Baglio F, Clerici M: Association between Hippocampal Shape, Neuroinflammation, and Cognitive Decline in Alzheimer’s Disease. J Alzheimers Dis 2018, 66(3):1131–1144. [DOI] [PubMed] [Google Scholar]
- 22.Janesick A, Wu SC, Blumberg B: Retinoic acid signaling and neuronal differentiation. Cell Mol Life Sci 2015, 72(8):1559–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Husson M, Enderlin V, Delacourte A, Ghenimi N, Alfos S, Pallet V, Higueret P: Retinoic acid normalizes nuclear receptor mediated hypo-expression of proteins involved in beta-amyloid deposits in the cerebral cortex of vitamin A deprived rats. Neurobiol Dis 2006, 23(1):1–10. [DOI] [PubMed] [Google Scholar]
- 24.Ding Y, Qiao A, Wang Z, Goodwin JS, Lee ES, Block ML, Allsbrook M, McDonald MP, Fan GH: Retinoic acid attenuates beta-amyloid deposition and rescues memory deficits in an Alzheimer’s disease transgenic mouse model. J Neurosci 2008, 28(45):11622–11634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nurrahmah QI, Madhyastha R, Madhyastha H, Purbasari B, Maruyama M, Nakajima Y: Retinoic acid abrogates LPS-induced inflammatory response via negative regulation of NF-kappa B/miR-21 signaling. Immunopharmacol Immunotoxicol 2021, 43(3):299–308. [DOI] [PubMed] [Google Scholar]
- 26.Qiao A, Li J, Hu Y, Wang J, Zhao Z: Reduction BACE1 expression via suppressing NF-kappaB mediated signaling by Tamibarotene in a mouse model of Alzheimer’s disease. IBRO Neurosci Rep 2021, 10:153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhong Y, Wu Y, Liu R, Li Z, Chen Y, Evans T, Chuang P, Das B, He JC: Novel retinoic acid receptor alpha agonists for treatment of kidney disease. PLoS One 2011, 6(11):e27945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Das BC, Nandwana NK, Das S, Nandwana V, Shareef MA, Das Y, Saito M, Weiss LM, Almaguel F, Hosmane NS et al. : Boron Chemicals in Drug Discovery and Development: Synthesis and Medicinal Perspective. Molecules 2022, 27(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ravelli KG, Rosario BD, Camarini R, Hernandes MS, Britto LR: Intracerebroventricular Streptozotocin as a Model of Alzheimer’s Disease: Neurochemical and Behavioral Characterization in Mice. Neurotox Res 2017, 31(3):327–333. [DOI] [PubMed] [Google Scholar]
- 30.Nasiri E, Sankowski R, Dietrich H, Oikonomidi A, Huerta PT, Popp J, Al-Abed Y, Bacher M: Key role of MIF-related neuroinflammation in neurodegeneration and cognitive impairment in Alzheimer’s disease. Mol Med 2020, 26(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Paladugu L, Gharaibeh A, Kolli N, Learman C, Hall TC, Li L, Rossignol J, Maiti P, Dunbar GL: Liraglutide Has Anti-Inflammatory and Anti-Amyloid Properties in Streptozotocin-Induced and 5xFAD Mouse Models of Alzheimer’s Disease. Int J Mol Sci 2021, 22(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Saito M, Smiley JF, Hui M, Masiello K, Betz J, Ilina M, Saito M, Wilson DA: Neonatal Ethanol Disturbs the Normal Maturation of Parvalbumin Interneurons Surrounded by Subsets of Perineuronal Nets in the Cerebral Cortex: Partial Reversal by Lithium. Cereb Cortex 2019, 29(4):1383–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fukasawa H, Nakagomi M, Yamagata N, Katsuki H, Kawahara K, Kitaoka K, Miki T, Shudo K: Tamibarotene: a candidate retinoid drug for Alzheimer’s disease. Biol Pharm Bull 2012, 35(8):1206–1212. [DOI] [PubMed] [Google Scholar]
- 34.Cramer PE, Cirrito JR, Wesson DW, Lee CY, Karlo JC, Zinn AE, Casali BT, Restivo JL, Goebel WD, James MJ et al. : ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models. Science 2012, 335(6075):1503–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dos Santos Guilherme M, Stoye NM, Rose-John S, Garbers C, Fellgiebel A, Endres K: The Synthetic Retinoid Acitretin Increases IL-6 in the Central Nervous System of Alzheimer Disease Model Mice and Human Patients. Front Aging Neurosci 2019, 11:182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Song S, Gao P, Sun L, Kang D, Kongsted J, Poongavanam V, Zhan P, Liu X: Recent developments in the medicinal chemistry of single boron atom-containing compounds. Acta Pharm Sin B 2021, 11(10):3035–3059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Coban FK, Ince S, Kucukkurt I, Demirel HH, Hazman O: Boron attenuates malathion-induced oxidative stress and acetylcholinesterase inhibition in rats. Drug Chem Toxicol 2015, 38(4):391–399. [DOI] [PubMed] [Google Scholar]
- 38.Behl T, Kaur D, Sehgal A, Singla RK, Makeen HA, Albratty M, Alhazmi HA, Meraya AM, Bungau S: Therapeutic insights elaborating the potential of retinoids in Alzheimer’s disease. Front Pharmacol 2022, 13:976799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nagamoto-Combs K, Kulas J, Combs CK: A novel cell line from spontaneously immortalized murine microglia. J Neurosci Methods 2014, 233:187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dave KM, Ali L, Manickam DS: Characterization of the SIM-A9 cell line as a model of activated microglia in the context of neuropathic pain. PLoS One 2020, 15(4):e0231597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Oliveira LM, Teixeira FME, Sato MN: Impact of Retinoic Acid on Immune Cells and Inflammatory Diseases. Mediators Inflamm 2018, 2018:3067126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Cai W, Wang J, Hu M, Chen X, Lu Z, Bellanti JA, Zheng SG: All trans-retinoic acid protects against acute ischemic stroke by modulating neutrophil functions through STAT1 signaling. J Neuroinflammation 2019, 16(1):175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.van Neerven S, Regen T, Wolf D, Nemes A, Johann S, Beyer C, Hanisch UK, Mey J: Inflammatory chemokine release of astrocytes in vitro is reduced by all-trans retinoic acid. J Neurochem 2010, 114(5):1511–1526. [DOI] [PubMed] [Google Scholar]
- 44.Goncalves MB, Clarke E, Hobbs C, Malmqvist T, Deacon R, Jack J, Corcoran JP: Amyloid beta inhibits retinoic acid synthesis exacerbating Alzheimer disease pathology which can be attenuated by an retinoic acid receptor alpha agonist. Eur J Neurosci 2013, 37(7):1182–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Leng F, Edison P: Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol 2021, 17(3):157–172. [DOI] [PubMed] [Google Scholar]
- 46.Kim RE, Shin CY, Han SH, Kwon KJ: Astaxanthin Suppresses PM2.5-Induced Neuroinflammation by Regulating Akt Phosphorylation in BV-2 Microglial Cells. Int J Mol Sci 2020, 21(19). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chen CH, Zhou W, Liu S, Deng Y, Cai F, Tone M, Tone Y, Tong Y, Song W: Increased NF-kappaB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int J Neuropsychopharmacol 2012, 15(1):77–90. [DOI] [PubMed] [Google Scholar]
- 48.Niu YL, Zhang WJ, Wu P, Liu B, Sun GT, Yu DM, Deng JB: Expression of the apoptosis-related proteins caspase-3 and NF-kappaB in the hippocampus of Tg2576 mice. Neurosci Bull 2010, 26(1):37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sun E, Motolani A, Campos L, Lu T: The Pivotal Role of NF-kB in the Pathogenesis and Therapeutics of Alzheimer’s Disease. Int J Mol Sci 2022, 23(16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhu H, Bai Y, Wang G, Su Y, Tao Y, Wang L, Yang L, Wu H, Huang F, Shi H et al. : Hyodeoxycholic acid inhibits lipopolysaccharide-induced microglia inflammatory responses through regulating TGR5/AKT/NF-kappaB signaling pathway. J Psychopharmacol 2022, 36(7):849–859. [DOI] [PubMed] [Google Scholar]
- 51.Boche D, Perry VH, Nicoll JA: Review: activation patterns of microglia and their identification in the human brain. Neuropathol Appl Neurobiol 2013, 39(1):3–18. [DOI] [PubMed] [Google Scholar]
- 52.Hu X, Li P, Guo Y, Wang H, Leak RK, Chen S, Gao Y, Chen J: Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke 2012, 43(11):3063–3070. [DOI] [PubMed] [Google Scholar]
- 53.Song GJ, Suk K: Pharmacological Modulation of Functional Phenotypes of Microglia in Neurodegenerative Diseases. Front Aging Neurosci 2017, 9:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Nagpal I, Wei LN: All-trans Retinoic Acid as a Versatile Cytosolic Signal Modulator Mediated by CRABP1. Int J Mol Sci. 2019, 20(15):3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lannert H, Hoyer S: Intracerebroventricular administration of streptozotocin causes long-term diminutions in learning and memory abilities and in cerebral energy metabolism in adult rats. Behav Neurosci 1998, 112(5):1199–1208. [DOI] [PubMed] [Google Scholar]
- 56.Mehla J, Pahuja M, Gupta YK: Streptozotocin-induced sporadic Alzheimer’s disease: selection of appropriate dose. J Alzheimers Dis 2013, 33(1):17–21. [DOI] [PubMed] [Google Scholar]
- 57.Salkovic-Petrisic M, Knezovic A, Hoyer S, Riederer P: What have we learned from the streptozotocin-induced animal model of sporadic Alzheimer’s disease, about the therapeutic strategies in Alzheimer’s research. J Neural Transm (Vienna) 2013, 120(1):233–252. [DOI] [PubMed] [Google Scholar]
- 58.Chen Y, Liang Z, Blanchard J, Dai CL, Sun S, Lee MH, Grundke-Iqbal I, Iqbal K, Liu F, Gong CX: A non-transgenic mouse model (icv-STZ mouse) of Alzheimer’s disease: similarities to and differences from the transgenic model (3xTg-AD mouse). Mol Neurobiol 2013, 47(2):711–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wang D, Wang C, Liu L, Li S: Protective effects of evodiamine in experimental paradigm of Alzheimer’s disease. Cogn Neurodyn 2018, 12(3):303–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hartlage-Rubsamen M, Zeitschel U, Apelt J, Gartner U, Franke H, Stahl T, Gunther A, Schliebs R, Penkowa M, Bigl V et al. : Astrocytic expression of the Alzheimer’s disease beta-secretase (BACE1) is stimulus-dependent. Glia 2003, 41(2):169–179. [DOI] [PubMed] [Google Scholar]
- 61.Lee DC, Rizer J, Selenica ML, Reid P, Kraft C, Johnson A, Blair L, Gordon MN, Dickey CA, Morgan D: LPS- induced inflammation exacerbates phospho-tau pathology in rTg4510 mice. J Neuroinflammation 2010, 7:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kiris E, Ventimiglia D, Feinstein SC: Quantitative analysis of MAP-mediated regulation of microtubule dynamic instability in vitro focus on Tau. Methods Cell Biol 2010, 95:481–503. [DOI] [PubMed] [Google Scholar]
- 63.Iqbal K, Liu F, Gong CX, Grundke-Iqbal I: Tau in Alzheimer disease and related tauopathies. Curr Alzheimer Res 2010, 7(8):656–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Doust YV, King AE, Ziebell JM: Implications for microglial sex differences in tau-related neurodegenerative diseases. Neurobiol Aging 2021,105:340–348. [DOI] [PubMed] [Google Scholar]
- 65.Mizobuchi H, Yamamoto K, Yamashita M, Inagawa H, Kohchi C, Soma GI: Prevention of streptozotocininduced Neuro2a cell death by C8B4 microglia transformed with repetitive lowdose lipopolysaccharide. Mol Med Rep 2021, 24(4). [DOI] [PubMed] [Google Scholar]
- 66.Biswas J, Goswami P, Gupta S, Joshi N, Nath C, Singh S: Streptozotocin Induced Neurotoxicity Involves Alzheimer’s Related Pathological Markers: a Study on N2A Cells. Mol Neurobiol 2016, 53(5):2794–2806. [DOI] [PubMed] [Google Scholar]
- 67.Ganz T, Fainstein N, Elad A, Lachish M, Goldfarb S, Einstein O, Ben-Hur T: Microbial pathogens induce neurodegeneration in Alzheimer’s disease mice: protection by microglial regulation. J Neuroinflammation 2022, 19(1):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
All data and materials are available in the article or upon request to corresponding authors.
