Design, drug-likeness and pharmacokinetic evaluation of 5,6-dimethoxy-indanone-
chalcone-carbamate hybrids as a novel MTDL
We designed and generated a new MTDL molecule (E)-3-((5,6-dimethoxy-1-oxo-1,3-
dihydro-2H-inden-2-ylidene)-methyl)phenylethyl(methyl)carbamate (named AP5) by binding 5,6-dimethoxy-indanoneand carbamate moiety with chalcone molecular scaffolds (Figure1a). Then, we first explored the binding potential of AP5 with the active pocket of AChE (PDB ID 2WHQ) through molecular docking. The molecular docking result showed that AP5 had moderate binding affinity with AChE binding pocket (Figure 1b) with lowest binding energy of -10.64 kcal/mol, compared to donepezil (-11.94kcal/mol) and rivastigmine (-8.55kcal/mol). The carbamate moiety of AP5 was buried deep into the catalytic site of AChE, however 5,6-dimethoxy
-indanone moiety contacted the peripheral sites at the edge of gorge (Figure 1b). Based on active amino acids in the vicinity of CAS and PAS of AChE [21], the binding of AP5 was stabilized by the hydrogen bond interaction of Phe295 and Arg296 of CAS, Tyr337 and Trp86 of PAS (Figure 1c). The other hydrophobic interaction was observed between 5,6-dimethoxy-indanone and side-chain Tyr124 located in PAS binding region. The carbamate group contacted the Trp286 and Leu289 in CAS region by hydrophobic interaction. AP5 also exhibited van der Waals interactions with Asp74, Gly120, Ser125 of PAS, and Ile294, Phe338 of CAS. Taken together, the docking analysis suggests that AP5 fits well in the active pocket of AChE by interacting with the peripheral and catalytic site residues simultaneously, which indicates its dual inhibition and high potency.
Combining several pharmacophores may lead to large molecule which may compromise drug-likeness properties [22]. Drug-likeness assessment in silicoby Lipinski's rule of five [23] showed that AP5 complied with the Lipinski's rule of five (Supplementary Table 1). Moreover, in silico assessment of blood-brain barrier (BBB) penetration showed that BBB-score >0.02 [24](Supplementary Table 1). These data indicate that AP5 holds appropriate drug-likeness properties and BBB permeability.
We next determine the pharmacokinetic profile of AP5 in male C57BL/6J mice given by a single intravenous (i.v.) dose of 10 mg/kg or single per os (p.o.) dose of 40 mg/kg, respectively. After a single oral dose, AP5 concentration in plasma reached the peak (2088 ng/mL) at 1.5 h by LC-MS/MS analysis. Importantly, we determined AP5 in the brain homogenate with the highest concentration of 95.3 ng/mL at 2h (Figure 1d-e). PK parameters revealed oral AP5 holds low systemic clearance (Cl =2.756 L/hr/kg), moderate volume of distribution (Vss=18.79 L/kg), highly favorable oral bioavailability F = 67.2% and adequate oral brain penetration (B/P = 10.23%) (Figure 1d-e,Supplementary Table 2). Taken together, AP5 showed adequate BBB permeability, suitable PK properties and drug-likeness properties, which supports the potential to develop AP5 as a promising candidate for studies in AD model.
AP5 inhibits AChE activity and blocks Aβ oligomerization or fibrillization
As result of carbamate pharmacophore of AP5 binding to CAS of AChE, we further examined the effects of AP5 on AChE activity in APP/PS1 mice. Consistent with previous study[25], our data showed that AChE activity in the cortex and hippocampus was markedly increased in APP/PS1 mice (Figure 2a), which led to reduction of ACh level compared to WT mice (Figure 2b). AP5-treated mice exhibited decreased activity of AChE compared with APP/PS1 mice in cortex (F2,15 = 7.730 p = 0.0049) and hippocampus (F2,15 = 29.21, p < 0.0001), and elevated levels of ACh in cortex (F2,15 = 7.886, p = 0.0046) and hippocampus (F2,15 = 22.51, p < 0.0001) (Figure 2a-b).
It is reported that Aβ could bind to AChE through PAS and induces its fibrillization [10, 11]. Propidium and fasciculin, PAS inhibitors, are capable of preventing the effect of AChE on Aβ fibril aggregation process [11]. As 5,6-dimethoxy-indanone moiety of AP5 binds AChE at PAS, we next evaluated whether AP5 exerted an inhibitory effects on Aβ fibrillogenesis induced by AChE. Firstly, we analyzed the inhibition of AP5 on Aβ fibril formation induced by AChE using 24h kinetic ThT fluorescence assay. As shown in Figure 2c, incubation of Aβ with AChE resulted in an increase in fluorescence intensity, confirming that AChE was able to promote Aβ assembly into fibrils. Conversely, co-incubation of AP5 and Aβ40with AChE substantially lowered the ThT intensity in a dose-dependent manner, indicating that AP5 inhibited the enhancement of Aβ fibrillization triggered by AChE. Besides, the effects of AP5 on disaggregation of pre-formed Aβ40 or Aβ42 fibrils was determined. Aβ40 or Aβ42 was firstly pre-aggregated with AChE for 2 days before co-incubation with AP5.In the presence of AP5, Aβ fibrils were disaggregated as shown by a marked decline of ThT fluorescence intensity in Aβ40 (F3,8 = 49.64, p < 0.0001) and Aβ42 (F3,12 = 43.32, p < 0.0001) (Figure 2d). This observation was further confirmed by dot blot assay with the amyloid fibrils-specific antibody (Aβ40, F4,10 = 93.92, p < 0.0001) (Aβ42, F4,10 = 161.9, p < 0.0001) (Figure 2e-f).
To study whether AP5 could inhibit Aβ self-oligomerization, dot blot assay was carried out with the amyloid oligomer-specific antibody. Monomeric Aβ40 or Aβ42 was incubated with or without AP5 under conditions that led to oligomerization. As indicated in Figure 2g-h, AP5 decreased the levels of oligomeric Aβ40 (F3,8 = 34.91, p < 0.0001) and Aβ42 (F3,8 = 13.92, p = 0.0015).This result was further corroborated by immunoblot detection that discriminated between Aβ oligomers and monomers. The synthetic oligomer preparations comprised a mixture of low molecular weight oligomers and high molecular weight (HMW) oligomers.HMW Aβ oligomers were observed as a smear on SDS-PAGE with typically ranging in molecular weight from 50-150 kDa [26]. As shown in Figure 2i, a 5 kDa Aβ monomer band, a 10 kDa Aβ dimer band as well as a smear were detected. In the presence of AP5, the intensity of the Aβ dimers and smear were decreased, demonstrating that AP5 specifically blocked Aβ oligomerization. Taken together, these data suggested that AP5 might not only elevated ACh level but also interfered toxic Aβ aggregation.
AP5 reduces Aβ deposition without altering APP processing in APP/PS1 mice
We next evaluated the overall effect of AP5 on Aβ pathology. Brain slices were stained with Thioflavin S (ThS) to detect dense-core fibrillar amyloid plaques. Indeed, we observed there was a pronounced reduction in the load (cortex, F2,5 = 86.47, p < 0.0001) (hippocampus, F2,15 = 44.26, p < 0.0001) and number (cortex, F2,15 = 246.6, p < 0.0001) (hippocampus, F2,15 = 150.8, p < 0.0001) of ThS-positive plaque in APP/PS1mice treated with AP5 (Figure 3a-b). Further this reduction was most pronounced for larger ( > 50μm2 in area) plaques in cortex (F2,15= 253.2, p < 0.0001) and hippocampus (F2,15 = 251.6, p < 0.0001). Similarly, immunofluorescent staining of Aβ peptide by MOAB-2 antibody, which recognizes the unaggregated, oligomeric and fibrillar form of Aβ, indicated that the area occupied by unaggregated and aggregated Aβ was also decreased in cortex (F2,15 = 82.42, p < 0.0001) and hippocampus (F2,15 = 64.06, p < 0.0001) of the AP5-administered AD mice(Figure 3c).
To further detect Aβ aggregation in these mice, we performed a serial extraction of cortex and hippocampus.TBS fraction contains more soluble Aβ species while the 70% FA fraction contains insoluble Aβ aggregates.APP/PS1 mice had significantly higher Aβ40 and Aβ42 levels in TBS- and FA-fractions (Figure 3d-e). Furthermore the majority of Aβ in APP/PS1 mice was fractioned in the FA-soluble fractions (Figure 3e), indicating its deposition in the amyloid plaque. In addition, as shown in Figure 3e, the concentrations of insoluble Aβ40 (cortex, F2,15 = 82.42, p < 0.0001) (hippocampus, F2,15 = 46.30, p < 0.0001) and Aβ42 (cortex, F2,15 = 132.4, p < 0.0001) (hippocampus, F2,15 = 112.2, p < 0.0001) were markedly decreased in FA-fractions, but no marked differences were determined in TBS-fraction after AP5 administration (Figure 3d).
Next, to clarity the mechanism that contributed to the reduction in Aβ following AP5 administration, we assessed APP processing by western blot. The results showed there wereno significant changes in the expression levels of the amyloid precursor protein (APP full length,APPfl), CTFs (C-terminal fragments, CTFβ), β-site-APP cleaving enzyme (BACE) and catalytic subunit of the gamma-secretase complex (presenilin 1), between APP/PS1 and AP5-treated APP/PS1 mice (Figure 5f-g). These data suggest that AP5 did not alter APP expression or processing. Overall, these results demonstrated that AP5 administration mitigates Aβ load without influencing Aβ production.
AP5 alleviates neuroinflammation of APP/PS1 mice
On the basis of the structure properties of AP5,which contains anti-inflammatory and antioxidative chalcone structure, we examined the effects of AP5 on neuroinflammation in APP/PS1 mice. Firstly, the brain slices were immunostained with ionized calcium-binding adapter molecule 1(Iba1) antibody and glial fibrillary acidic protein (GFAP) to investigate the effects of AP5 on microgliosis and astrogliosis, respectively. We morphologically classified Iba1+ microglia to quantify microglia activation state, and observed that microglial cells in APP/PS1 mic eexhibited a significantly elevated proportion of intermediate, amoeboid or round, showing an increase in microglia activation. For the APP/PS1 mice treated with AP5, we found a marked decrease in the proportion of activated phenotype (F2,15 = 10.71, p = 0.0013) (Figure 4a-b). Consistent with previous study [27], quantitative analysis of individual reactive astrocyte morphology using Sholl analysis showed that elevated ramification index (F2,15 = 44.12, p < 0.0001) and the sum of intersects (F2,15 = 82.97, p < 0.0001) in APP/PS1 mice. Similar to microglia, these reactive phenotypes of astrocytes were attenuated by AP5 treatment (Figure 4c-d). However, the ending radius, an indicator of astrocyte territory, was not changed in APP/PS1 mice.
Next, we determined the levels of several proinflammatory cytokines (TNF-α, IL-6 and IL-1β) implicated in AD. A greatly increased secretion of TNF-α (F2,15 = 107.0, p < 0.0001), IL-6 (F2,15 = 54.31, p < 0.0001) and IL-1β (F2,15 = 25.01, p < 0.0001) were detected in APP/PS1 mice, which were markedly reduced by AP5 administration (Figure 4e).Together, these results show AP5 attenuated neuroinflammatory responses in AD mice model.
It has been reported that microglia clustered in the vicinity of Aβ plaque form a protective barrier which compacts amyloid fibrils and reduces their toxicity [28]. Thus, we quantified the Aβ plaque-associated microglia by immunofluorescent co-staining with MOAB-2 and Iba1.The increased clustering of microglia surrounding a plaque was observed in brains of AP5-treated APP/PS1 mice (Figure 4f-g). Microglia clustering around plaques facilitate extracellular Aβ clearance [29]. Thus, we further investigated whether the increased microglia recruitment in the vicinity of plaques upon AP5 treatment can promote Aβ phagocytosis. As shown in Figure 4j-k, the area of CD68 (a marker of phagocytic activity of microglia) within microglia was increased, suggesting that AP5 treatment enhances microglial response to Aβ plaques and phagocytic activity. These results revealed that AP5 decreased microglia activation and enhanced the clustering of microglia around Aβ plaque, thereby diminishing plaque-associated neurotoxicity and promoting microglia phagocytosis and clearance of Aβ. Besides, we found there was no statistically difference in the number of astrocytes around Aβ plaque (Figure 4h-i).
AP5 rescues the neuron and synapse damage in the APP/PS1 mice
Aβ aggregation in the brain is a key initiating step in the pathogenesis of AD, which induces many downstream detrimental events such as oxidative stress, neuroinflammation, synaptic impairment, neuronal degeneration, and eventual cognitive defects [2]. Therefore, we next examined whether the protective effects of AP5 against Aβ-induced neurotoxicity was reflected at the neuronal and synaptic level. Firstly, to determine the effects of AP5 on neuron loss and degeneration in APP/PS1 mice, we carried out co-immunostaining with NeuN and MAP2. Remarkably, we detected that the average number of NeuN+ cells was reduced (F2,15 = 21.55, p < 0.0001), and MAP2 signals were also significantly disrupted (F2,15 = 30.17, p < 0.0001) in hippocampal CA1 pyramidal layer of APP/PS1 mice, but these changes were restored in AP5-treated APP/PS1 mice (Figure 5a-b). Consistent with these results, the protein expression of NeuN and MAP2 was markedly increased in hippocampus of AP5-treated mice (Supplementary Figure 2a-b).
Next, we investigated the efficacy of AP5 against synaptic pathology in APP/PS1 mice.
synaptophysin and PSD95 are two membrane protein markers located in the presynaptic and postsynaptic cells, respectively. Co-immunofluorescence of synaptophysin with PSD95 revealed reduced pre- (F2,15 = 16.1, p = 0.002), post- (F2,15 = 20.04, p < 0.0001), and double-positive
-synaptic puncta (F2,15 = 37.82, p < 0.0001) in APP/PS1 mice, and the synapse damage was markedly recovered by AP5 (Figure 5c-d).Western blot further indicated that the expression levels of synaptophysin and PSD95 in hippocampus of AP5-treated mice were more than that in the APP/PS1 mice, respectively (Supplementary Figure 2a-b).
These results suggested that AP5 prevented or reversed neuronal and synaptic damage, which might play a pivotal role in ameliorating downstream cognitive defects.
Chronic dosing AP5 from the early stages of β-amyloidosis alleviates cognitive deficits of APP/PS1 mice
We next assessed whether the beneficial effects of AP5 in decreasing Aβ plaque deposition and neuroinflammation are accompanied by cognitive improvement of APP/PS1 mice. Firstly, we evaluated general locomotor activity of WT, APP/PS1 and AP5 mice through the open field test, and no difference in the total distance traveled (F2,27 = 0.6091, p = 0.5511) was observed among groups, indicating that AP5 administration did not elicit any influence on general locomotor activity of the mice (Figure 5e). Furthermore, we employed Y-maze to evaluate spatial recognition memory. AP5-treated mice demonstrated a marked elevation of their preference index (F2,27 = 11.07, p = 0.0003) relative to APP/PS1 mice after 3 h ’s training, suggesting the improvement of short-term spatial recognition memory (Figure 5f).
Moreover, we used the novel object recognition task and the spatial object location task to evaluate recognition memory. APP/PS1 mice demonstrated impaired short- and long-term recognition memory by a significant decline of its discrimination index at 1h and 24h compared with WT mice (Figure 5g-h). AP5-treated APP/PS1 mice performed significantly better than APP/PS1 mice without treatment in the novel object recognition task (F2,27 = 8.215, p = 0.0016) and the spatial object location task (F2,27 = 11.05, p = 0.0003) at 24h (Figure 5g-h). In addition, the discrimination index in AP5-treated AD mice for novel object recognition was significantly higher (F2,27 = 9.417, p = 0.0008) than APP/PS1 mice at 1h (Figure 5g). These data indicate an improvement of short- and long-term recognition memory upon AP5 treatment.
We examined the reference memory through Morris water maze (MWM). As indicated in Figure 5i, APP/PS1 mice exhibited marked spatial learning and memory decline by longer escape latency in the successive platform learning trials, less time spent in the target quadrant and lower platform crossover number in the probe trail (Figure 5k). The swimming velocity during the successive training period were indistinguishable (F2,27 = 0.7216, p = 0.4951) between each group (Figure 5j). After 5 months’ treatment of AP5, the mice performed much better in escape latency including the time effect (F2.797,75.51= 66.46, p < 0.0001) and group effect (F2,27= 79.06, p < 0.0001) by two-way ANOVA, platform crossing number (F2,27 = 8.695, p = 0.0012) and time spent in the target quadrant (F2,27 = 9.236, p = 0.0009) (Figure 5k), suggesting that AP5 slows cognitive decline in AD mice.
AP5 regulates gene transcription in APP/PS1 mice
Given the multitargeted effects of AP5 that we observed, we next explore the underlying molecular mechanism by high-throughput bulk RNA sequencing (RNA-seq) of the hippocampal tissues. A total of 17981 genes were identified in the hippocampi from above WT, APP/PS1 and AP5 mice. Volcano plots depicted that 933 genes were differentially expressed, of which 425 genes were induced and 508 genes were suppressed in APP/PS1 mice treated with AP5 treatment compared with APP/PS1 mice without treatment (Figure 6a). Gene ontology (GO) term analysis demonstrated that, in addition to regulation of gene expression, inflammation-related features were enriched, including microglial activation involved in immune response, astrocyte activation, synapse pruning and complement-mediated synapse pruning as well as processes responsible for phagocytosis, engulfment and Aβ clearance (Figure 6b). In agreement with previous reports[30], deposition of Aβ in AD patients is associated with the innate immune system activation. Together, these data suggested that AP5 is of great importance to regulate inflammation in disease progression.
Because microglia and astrocyte are the main cell types exerting inflammatory effects in the brain, we carried out heatmaps of disease-associated microglia (DAM) genes from hippocampal RNA-seq data. We analyzed gene expression profile in the hippocampus and observed an increase in DAM gene expression in APP/PS1 mice with AP5 treatment compared with APP/PS1 mice without treatment (Figure 6c), further suggesting that microglial cells acquired a typical DAM phenotype. The ability of microglia to switch to a DAM phenotype appears essential to limit Aβ plaque deposition and inflammation, thereby removing the neuronal damage in AD [31]. Moreover, CD11c (encoded by Itgax) has recently been identified as a marker for DAM [31]. We found CD11c-positive microglia were accumulated around Aβ plaques. Notably, CD11c-positive area within activated microglia was significantly elevated with AP5 treatment (Figure 6d-e), further confirming that AP5 skewed microglia to a DAM phenotype and contributed to the decreased deposition of Aβ and inflammation.
At the same time, we compared the transcriptional profile of reactive astrocytes with that of astrocytes subpopulations recently identified pan-reactive, A1 and A2 specific astrocytes [32]. A1 specific astrocytes, induced by neuroinflammation, are powerfully neurotoxic, where their presence may well account for neurodegeneration and drive disease deterioration. However, A2 astrocytes upregulate many neurotrophic factors, and therefore are protective. As expected, astrocytes in the hippocampus of APP/PS1 mice displayed an increased gene expression of pan-reactive and A1specific astrocytes signatures. AP5 reduced the expression of most genes linked to pan-reactive and A1 astrocytes (Figure 6f), suggesting that AP5 could prevent or revert A1-specific astrocytes formation. No differences among groups were detected in the expression of signature genes of A2-specific astrocytes. Since complement C3 (C3) is one of the most distinctive and highly elevated genes in A1 specific astrocytes and is not expressed by A2 specific astrocytes [33], we performed immunofluorescence to identify whether C3-expressing A1 astrocytes are decreased following AP5 treatment. We found C3-positive area within astrocytes was markedly reduced after AP5 treatment (Figure 6g-h), which further confirming AP5 could prevent and reverse A1specific astrocyte formation. Taken together, our data demonstrated that regulation of astrocyte and microglia phenotype transition by AP5 may be an important mechanism to control inflammation in AD.