Next Article in Journal
Anion-Complexation-Induced Emission Based on Aggregation-Induced Emission Fluorophore
Previous Article in Journal
An Update on Phytochemicals in Redox Homeostasis: “Virtuous or Evil” in Cancer Chemoprevention?
Previous Article in Special Issue
Resveratrol Analogues as Dual Inhibitors of Monoamine Oxidase B and Carbonic Anhydrase VII: A New Multi-Target Combination for Neurodegenerative Diseases?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Homotaurine and Curcumin Analogues as Potential Anti-Amyloidogenic Agents

1
School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
2
School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA
*
Author to whom correspondence should be addressed.
Chemistry 2023, 5(1), 223-241; https://doi.org/10.3390/chemistry5010018
Submission received: 24 July 2022 / Revised: 13 January 2023 / Accepted: 6 February 2023 / Published: 7 February 2023

Abstract

:
Currently, there is neither a cure for Alzheimer’s disease (AD) nor a way to stop the progressive death of neuronal cells associated with this devastating aliment. To date, there are only medications that temporarily slow its progression, and do not interfere with its pathogenesis. One of the hallmarks of AD is the presence of amyloid-beta plaques derived from the metabolism of the amyloid precursor protein, via the cleavage by beta followed by gamma secretase. Homotaurine, a naturally occurring small molecule found in some seaweeds, and curcumin, a phenolic antioxidant found in Curcuma longa, have been extensively studied as potential compounds to prevent/reverse plaque formation. In this study, libraries of chalcones and extended chalcones based on curcumin, as well as aminopropylsulfonamides inspired by homotaurine, were synthesized. Using fluorescence spectroscopic analysis with Thioflavin T, the anti-aggregation effect on Aβ42 was determined. A select number of newly synthesized chalcones and extended chalcone analogs were revealed to be potential anti-amyloidogenic agents. These were further evaluated with regard to their neurotoxicity/neuroprotection. The extended chalcone analogs that displayed the most anti-aggregation effect on Aβ42 were further analyzed in an MTT assay. Although none of the compounds alone displayed any neurotoxicity, none were able to provide neuroprotection against Aβ42.

1. Introduction

Alzheimer’s disease (AD), the most prominent form of dementia, is an irreversible multifaceted, progressive brain disorder that displays slow cognitive decline [1]. The etiology of AD is not fully understood, however, studies have demonstrated that AD is related to low levels of acetylcholine [2], aggregates of amyloid-beta peptide (Aβ) [3], hyperphosphorylation of tau proteins [4], and oxidative stress [5]. The amyloid cascade hypothesis suggested that a sequence of abnormalities in the cleavage of the amyloid precursor protein (APP) [6] leads to the production and accumulation of insoluble Aβ peptides, with the most common isoforms being 40 (Aβ40) or 42 (Aβ42) amino acids in length [7]. The Aβ peptides can aggregate into oligomers, forming insoluble fibrils, ultimately leading to plaques. These plaques interfere in neuronal signaling which disrupts brain cell functions. The result is a loss of neuronal synapses, progressive decline in neurotransmitter activity, inflammation, and neuronal cell death. The amyloid cascade hypothesis suggests that the Aβ plaques initiate mitochondrial oxidative stress and promote hyperphosphorylation of tau proteins, resulting in neurotoxicity [8]. Amyloid inhibitor therapies have been attempted to reduce Aβ peptide production, either via: α-secretase stimulation, inhibition of γ-secretase, and/or inhibition of β-secretase [9]. Earlier this year, aducanumab, a monoclonal antibody developed by Biogen which targets the Aβ aggregates, was approved by the FDA and became the first drug to treat AD [10].
Homotaurine (Figure 1) is a natural product found in marine red algae and has been chemically synthesized [11]. It was clinically tested through Phase 3, as a potential anti-amyloidogenic agent for AD [12] Unfortunately, it failed to show statistically significant superiority over the placebo [12]. Curcumin (Figure 1), an orange-yellow polyphenol found in turmeric, underwent clinical study in 2008. A six-month randomized, placebo-controlled, double-blind, clinical trial failed to show health benefits [13]. It was suggested that curcumin may act on AD by Aβ disaggregation, anti-inflammation, and/or antioxidation [13]. In vitro studies revealed that curcumin inhibited Aβ40 aggregation and prevented Aβ42 oligomer formation at concentrations between 0.1 and 1.0 μM but may have BBB permeability issues [14]. We thus aimed to design a series of sulfonamides and chalcone derivates based upon the structures of homotaurine and curcumin, respectively, and determined their activity as possible anti-amyloidogenic agents.

2. Results and Discussion

2.1. Homotaurine-Based Analogues

We hypothesized that homotaurine’s lack of clinical success may have been related to the highly anionic nature and subsequent low logP. The low logP was attributed to possessing both sulfonic acid and amine moieties, thus hindering membrane permeability. Thus, we looked at replacing this moiety with a weak acid, specifically sulfonamide. We synthesized sulfonamide derivatives from 3-chloropropanesulfonyl chloride (Scheme 1) whereby several primary and secondary amines could be employed. It was crucial that we utilized a primary amine in the first step to ensure the presence of an acidic proton in the final sulfonamide product. The reaction of sulfonyl chloride with various amines was carried out in THF at 0 °C, as this was a highly exothermic reaction. In the second step, it was determined that the addition of KI facilitated the nucleophilic attack on the alkyl chloride, reducing the reaction time from >96 to 48 h. We avoided using water to allow for easier purification of this potential zwitterion via flash chromatography. The final chemical yields ranged from 9 to 71%, primarily driven by sterics of the amine.
The in vitro anti-aggregation of the amyloid-beta (142) peptide was conducted with all compounds initially at 100 µM and incubated at 37 °C, in the presence of Thioflavin T (200 µM) over 120 min. Homotaurine itself did not significantly affect the aggregation, while our positive control (Phenol red) showed a significant decrease over 2 h (Figure 2). Unfortunately, none of the sulfonamide derivatives showed any noticeable effect on the aggregation of the Aβ peptide. In fact, the kinetic curves of all synthesized sulfonamides were similar to the homotaurine and, at the same time, similar to the peptide without the inhibitor (Figure 2), thus, the IC50 values could only be estimated as >100 μM (Table 1).

2.2. Curcumin-Based Analogues

With knowledge that curcumin has been reported to display anti-amyloidogenic activity [14], we synthesized nine chalcones (14–22) in addition to two hybrid chalcone-sulfonamide derivatives (23, 24). Most of the chalcones were synthesized via a classic condensation coupling of an acetophenone and a benzaldehyde (Scheme 2). This one-step synthesis yielded our products without the need for further purification. Due to the fact that the final products were not soluble in the cold water/ethanol solution, while the starting materials were, simple filtration and washing were utilized. A few chalcones (21–24) were synthesized by using boron trifluoride etherate (BF3.Et2O) as a condensing agent in the reaction. This BF3.Et2O-assisted method produces higher chemical yields, requiring shorter reaction times, with minimal side reactions [15].
We again tested each of the chalcones in the same ThT assay, initially at 100 μM for 120 min. Modest activity for the majority of the chalcones possessing a 4-dimethylamino group on ring B was observed, with a kinetic curve being similar to Phenol red (Figure 3). The chalcone-sulfonamide hybrid compounds (23,24) again displayed a lack of activity towards Aβ aggregation. Therefore, we abandoned any further homotaurine analogs.
Curcumin’s extended conjugation (specifically in its enol tautomeric form) inspired us to extend our chalcone derivatives. We synthesized a diversity of extended chalcones with different substitution patterns as well as the inclusion of some with fused ring systems (31–33) via the condensation of an acetophenone and a cinnamaldehyde (Scheme 3). A noticeable improvement in anti-amyloidogenic profiles for those possessing a 4-dimethylamino group on ring B was observed (Figure 4).
All chalcones (1424) and extended chalcones (2535) were initially screened at 100 μM. Active compounds were defined as those displaying a kinetic curve, similar to or superior to Phenol red. These were further tested to obtain IC50 values (determined by linear regression parameter), again utilizing the ThT assay [16,17,18]. Encouragingly, four chalcones (1618, and 20) inhibited Aβ peptide aggregation with IC50 values in the micromolar range (Table 2). Moreover, five extended chalcones (26,27,29,34,35) also displayed noticeable anti-amyloidogenic activity (Table 2)—the most potent being compound 26 from the extended chalcone library with an IC50 value of 2.43 µM. Again, generally the most active compounds possessed the 4-dimethylamino on ring B. Overall, it was clear that the extended chalcones displayed superior activity over the simple chalcones with the same substituent pattern on the aromatic rings. For example, 20 had an IC50 value of 40.2 µM, compared to the extended chalcone counterpart (i.e., 34) of 18.7 µM. This was also observed with 17 compared to 27 (6.9 vs. 3.4 µM, respectively) and 16 compared to 26 (66.2 vs. 2.4 µM, respectively). Other electron-donating (OMe) or -withdrawing groups (CN, Cl, or NO2) on ring B for either the chalcone or extended chalcone showed no activity. Conversely, both electron-donating and -withdrawing groups (Cl, Me, CF3) or fused rings (29 and 35) on ring A displayed good anti-amyloidogenic activity as long as the 4-dimethylamino on ring B was present.
In addition, we wanted to determine whether the compounds showing low IC50 values, (26, 27, and 34) exhibited any neuroprotection using a neuronal cell line; specifically, neuroblastoma SH-SY5Y cells. The cells were obtained from the neuroblastoma cell line SK-N-SH and maintained in DMEM:F12 media. Of note, a plethora of cytotoxicity assays have been reported; however, these are not necessarily useful in these studies for a variety of reasons. For example, the lactate dehydrogenase (LDH) release assay was attempted [19]; unfortunately, we did not obtain reportable data from this study. This was due to the fact that the LDH release assay assesses necrotic cell death, whereas amyloid beta causes apoptotic cell death in SH-SY5Y cells. We next performed a CellTiter-Glo assay [20], which determines the number of viable cells by quantifying the amount of ATP. This assay did not produce positive data since ATP is required for amyloid-beta-induced apoptosis. The MTS assay [21] also did not yield any significant data. Ultimately, the cell viability/cell cytotoxicity-based 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed according to the previous literature (Figure 5) [22]. The cell viability of SH-SY5Y cells exposed to compounds 26, 27, and 34 were measured at their IC50 values. All three compounds displayed no significant changes in cell viability at these concentrations. Conversely, the addition of Aβ42 led to a ~40% decrease in cell viability, highlighting its known neuro-cytotoxicity. Unfortunately, the co-administration of compounds 26, 27, or 34 at their IC50 concentration did not demonstrate significant decreases in Aβ42-induced cytotoxicity in SH-SY5Y cells.

3. Conclusions

Unfortunately, the synthesized homotaurine-inspired library displayed no anti-amyloid genic activity even at very high concentrations. However, some of our curcumin-based analogous, whether chalcones or extended chalcones, did display activity against Aβ aggregation. Those with an electron-donating group, specifically 4-dimethylamino on the B side, displayed the greatest activity. Currently, it is unclear whether electron-withdrawing or -donating groups on the A side increases activity. When neurotoxicity studies were performed on the three extended chalcones with the greatest activity, no notable neuroprotection was observed. Future studies will focus upon ascertaining what modifications are necessary to translate positive ThT assay outcomes into cell viability results.

4. Materials and Methods

All chemicals were purchased from Millipore Sigma and used without further purification. All synthesized compounds were purified using flash column chromatography. 1H-NMR and 13C-NMR were recorded at 300 MHz on a Varian instrument using VnmrJ version 4.2A. NMR spectroscopy for compounds 21–24 was performed on a Bruker AVANCE III 400 MHz spectrometer. For in vitro studies, a fluorometric assay was performed in 96 non-binding microplates from Greiner Bio-One with a clear bottom on a Synergy Bio-tek HTS plate reader.

4.1. Preparation and Characterization of Homotaurine and Curcumin Analogues

  • N-(isobutyl)-3-(isobutylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (0.89 g, 5.03 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isobutyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and isobutyl amine (0.89 mL, 8.95 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.52 g, 42%). 1H NMR (300 MHz, CDCl3) δ 0.90 (d, J = 6.7 Hz, 6H), 0.95 (d, J = 6.7 Hz, 6H), 1.76 (m, 2H), 1.98 (quint, J = 6.5 Hz, 1H), 2.40 (d, J = 6.8 Hz, 2H), 2.73 (t, J = 6.4 Hz, 2H), 2.91 (d, J = 6.8 Hz, 2H), 3.11 (t, J = 7.3 Hz, 2H). 13C NMR (75 MHz, DMSO-d) δ 54.91, 50.25, 48.82, 46.53, 28.86, 26.25, 21.18, 20.68, 20.40.
2.
N-(isobutyl)-3-(diethylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (0.58 g, 3.27 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isobutyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and diethyl amine (0.42 mL, 5.7 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.15 g, 18%). 1H-NMR (300 MHz, CDCl3) δ 0.95 (d, J = 6.7 Hz, 6H), 1.00 (t, J = 7.2 Hz, 6H), 1.78 (m, 1H), 1.94 (quint, J = 7.4 Hz, 2H), δ 2.52 (m, 6H), δ 2.90 (t, J = 5.2 Hz, 2H), 3.07 (t, J = 7.6 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 51.14, 50.72, 50.54, 46.48, 29.00, 21.54, 19.89, 11.42.
3.
N-(t-butyl)-3-(diethylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.52 g, 8.57 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby t-butyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and diethyl amine (2 mL, 19.2 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (1.00 g, 46%). 1H-NMR (300 MHz, CDCl3) δ 0.98 (t, J = 7.15 Hz, 6H), 1.36 (s, 9H), 1.92 (quint, J = 7.2 Hz, 2H), 2.5 (m, 6H), 3.06 (t, J = 7.7 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 54.47, 54.40, 51.05, 46.56, 30.34, 22.00, 11.61.
4.
N-butyl-3-(diethylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.46 g, 8.22 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby n-butyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and diethyl amine (0.42 mL, 5.7 mmol) were added and the mixture was heated to 130 °C for 48 hrs. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (1.46 g, 71%). 1H-NMR (300 MHz, CDCl3) δ 0.91 (t, J = 7.4 Hz, 3H), 0.98 (t, J = 7.1 Hz 6H), 1.36 (m, 2H), 1.52 (quint, J = 7.8 Hz, 2H), δ 1.92 (quint, J = 7.2 Hz, 2H), 2.50 (m, 6H), 3.06 (m, 4H). 13C-NMR (75 MHz, CDCl3) δ 51.23, 50.84, 46.46, 42.93, 32.41, 21.63, 19.78, 13.63, 11.46.
5.
N-isopropyl-3-(diethylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (0.59 g, 3.33 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isopropyl amine (4 mL, 0.05 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and diethyl amine (1 mL, 9.6 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.55 g, 70%). 1H-NMR (300 MHz, CDCl3) δ 1.02 (d, J = 7.4 Hz, 6H), δ 2.04 (quint, J = 7.2 Hz, 2H), 2.93 (t, J = 7.6 Hz, 2H), 3.35 (m, 1H), 3.5 (m, 4H).
6.
N-isobutyl-3-(dipropylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.30 g, 7.32 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isobutyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and dipropyl amine (2 mL, 14.62 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.41 g, 20%). 1H-NMR (300 MHz, CDCl3) δ 0.86 (t, J = 7.3 Hz, 6H), 0.95 (d, J = 6.7 Hz, 6H), 1.42 (sextet, J = 7.4 Hz, 4H), 1.79 (m, 1H), 1.92 (m, 2H), 2.35 (t, J = 7.4 Hz, 4H), 2.51 (t, J = 6.5 Hz, 2H), 2.92 (t, J = 6.3 Hz, 2H), 3.08 (t, J = 7.6 Hz, 2H). Yield: 70%. 13C-NMR (75 MHz, CDCl3) δ 55.83, 52.41, 50.84, 50.61, 29.01, 21.84, 20.10, 19.88, 11.91.
7.
N-isopropyl-3-(dipropylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.01 g, 5.70 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isopropyl amine (4 mL, 0.05 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and dipropyl amine (2 mL, 14.62 mmol) were added and the mixture was heated to 130 °C for 48 hrs. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.22 g, 14%). 1H-NMR (300 MHz, CDCl3) δ 0.82 (t, J = 7.4 Hz, 6H), 1.19 (d, J = 6.5 Hz, 6H), 1.39 (m, 4H), 1.88 (quint, J = 6.8 Hz, 2H), 2.32 (t, J = 7.5 Hz, 4H), 2.47 (t, J = 6.4 Hz, 2H), 3.04 (t, J = 7.8 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 55.68, 52.16, 51.94, 46.00, 24.19, 21.73, 19.99, 11.83, 11.81, 11.79.
8.
N-butyl-3-(dipropylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.31 g, 7.39 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby n-butyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and dipropyl amine (2 mL, 14.62 mmol) were added and the mixture was heated to 130 °C for 48 hrs. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.19 g, 10%). 1H-NMR (300 MHz, CDCl3) δ 0.85 (t, J = 7.3 Hz, 6H), 0.91 (t, J = 7.3 Hz, 3H), 1.40 (m, 6H), 1.53 (quint, J = 7.6 Hz, 2H), 1.92 (quint, J = 7.5 Hz, 2H), 2.36 (t, J = 7.5 Hz, 4H), 2.52 (t, J = 6.5 Hz, 2H), 3.07 (m, 4H). 13C-NMR (75 MHz, CDCl3) δ 55.64, 52.27, 50.68, 42.98, 32.39, 21.67, 19.89, 19.79, 13.64, 11.90.
9.
N-(t-butyl)-3-(dipropylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.33 g, 7.52 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby t-butyl amine (4 mL, 0.04 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and dipropyl amine (2 mL, 14.62 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.74 g, 35%). 1H-NMR (300 MHz, CDCl3) δ 0.86 (t, J = 7.3 Hz, 6H), 1.41 (m, 13H), 1.92 (quint, J = 6.8 Hz, 2H), 2.35 (t, J = 7.5 Hz, 4H), 2.50 (t, J = 6.8 Hz, 2H), 3.10 (t, J = 7.8 Hz, 2H), 4.32 (s, 1H). 13C-NMR (75 MHz, CDCl3) δ 55.86, 54.44, 54.40, 52.31, 30.33, 22.14, 20.17, 11.91.
10.
N-isopropyl-3-(butyl(ethyl)amino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.43 g, 8.07 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isopropyl amine (4 mL, 0.05 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and N-ethylbutyl amine (2 mL, 16.75 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate:hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.22 g, 10%). 1H-NMR (300 MHz, CDCl3) δ 0.92 (t, J = 7.5 Hz, 3H), 1.04 (t, J = 7.0 Hz, 3H), 1.24 (d, J = 6.5 Hz, 6H), 1.29 (m, 2H), 1.44 (m, 2H), 1.99 (quint, J = 7.2 Hz, 2H), 2.48 (t, J = 7.8 Hz, 2H), 2.60 (m, 4H), 3.08 (t, J = 7.5 Hz, 2H), δ 3.64 (quint, J = 6.5 Hz, 1H). 13C-NMR (75 MHz, CDCl3) δ 52.70, 52.03, 51.46, 47.01, 46.18, 28.49, 24.34, 21.46, 20.61, 14.02, 11.09.
11.
N-benzyl-3-(t-butylamino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.22 g, 6.89 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby benzyl amine (4 mL, 0.03 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and t-butyl amine (2 mL, 19.03 mmol) were added and the mixture was heated to 130 °C for 48 hrs. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate:hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (1.07 g, 54%). 1H-NMR (300 MHz, CDCl3) δ 1.01 (s, 9H), 1.92 (quint, J = 6.8 Hz, 2H), 2.63 (t, J = 6.2 Hz, 2H), 3.07 (t, J = 6.2 Hz, 2H), 3.69 (s, 1H), 4.28 (d, J = 7.8 Hz, 2H), 7.32 (m, 5H).
12.
N-isopropyl-3-(butyl(methyl)amino)propane-1-sulfonamide
3-chloropropanesulfonyl chloride (0.88 g, 4.96 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isopropyl amine (4 mL, 0.05 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and N-methylbutyl amine (1.6 mL, 12.66 mmol) were added and the mixture was heated to 130 °C for 48 h. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate:hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.16 g, 9%). 1H-NMR (300 MHz, CDCl3) δ 0.89 (t, J = 7.1 Hz, 3H), 1.21 (d, J = 6.5 Hz, 6H), 1.28 (m, 2H), 1.41 (m, 2H), 1.94 (m, 2H), 2.18 (s, 3H), 2.31 (t, J = 7.4 Hz, 2H), 2.42 (t, J = 6.6 Hz, 2H), 3.06 (t, J = 7.6 Hz, 2H), 3.61 (quint, J = 6.4 Hz, 1H). 13C-NMR (75 MHz, CDCl3) δ 57.34, 55.86, 52.18, 46.07, 41.79, 29.25, 24.36, 21.74, 20.62, 14.06.
13.
N-isopropyl-3-morpholinopropane-1-sulfonamide
3-chloropropanesulfonyl chloride (1.44 g, 8.15 mmol) was added to a round-bottom flask at 0 °C and dissolved in THF (3 mL) under argon, whereby isopropyl amine (4 mL, 0.05 mol) was added dropwise. The mixture was stirred for 20 min, after which time THF and the excess of amine were evaporated under reduced pressure. The remaining residue was dissolved in dichloromethane (10 mL) and washed with 1 M hydrochloric acid (10 mL). The organic layer was dried over magnesium sulfate, filtered, and evaporated. 1H-NMR was run on this intermediate to confirm complete conversion and used without further purification. The resulting liquid product was transferred to a pressure vessel and dissolved in toluene (4 mL). Potassium iodide (10 mg, 0.06 mmol) and morpholine (1 mL, 11.49 mmol) were added and the mixture was heated to 130 °C for 48 hrs. The resulting yellow liquid was filtered and the excess of toluene was evaporated. The resulting mixture was purified using flash column chromatography (gradient elution-ethyl acetate: hexane with an increase in ethyl acetate from 66% to 100%) to obtain a yellow liquid (0.16 g, 32%). 1H-NMR (300 MHz, CDCl3) δ 1.20 (d, J = 6.7 Hz, 6H), 1.95 (quint, J = 7.5 Hz, 2H), 2.41 (m, 6H), 3.06 (t, J = 7.7 Hz, 2H), 3.59 (m, J = 6.7 Hz, 1H), 3.67 (t, J = 4.6 Hz, 4H), 4.67 (d, J = 7.6 Hz, 1H). 13C-NMR (75 MHz, CDCl3) δ 66.84, 56.76, 53.41, 51.90, 46.12, 24.34, 20.87.
14.
(E)-Chalcone
A solution of acetophenone (1.05 g, 8.17 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0 °C. The mixture was stirred for 15 min, after which time benzaldehyde (1.0347 g, 9.75 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as a white solid (1.52 g, 75%). 1H-NMR (300 MHz, CDCl3) δ 7.42 (m, 3H), 7.54 (m, 4H), 7.65 (m, 2H), 7.82 (d, J = 15.7 Hz, 1H), 8.02 (d, J = 9 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 190.59, 144.88, 138.20, 134.87, 132.82, 130.58, 128.98, 128.65, 128.52, 128.47, 122.06. Melting point: 52.6–55.3 °C.
15.
(E)-1-(4-methoxyphenyl)-3-phenylprop-2-en-1-one
A solution of 1-(4-methoxyphenyl)ethan-1-one (1.02 g, 6.82 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0 °C. The mixture was stirred for 15 min, after which time benzaldehyde (1.0324 g, 9.72 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as a white solid (1.87 g, 81%). 1H-NMR (300 MHz, CDCl3) δ 3.89 (s, 3H), 6.98 (d, J = 8.9 Hz, 2H), 7.42 (m, 3H), 7.55 (d, J = 15.7 Hz, 1H), 7.64 (m, 2H), 7.80 (d, J = 15.7 Hz, 1H), 8.05 (d, J = 8.9 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 188.74, 163.43, 143.99, 135.07, 130.83, 130.35, 128.93, 128.37, 125.76, 121.86, 113.85, 55.52. Melting point: 104.0–107.4 °C.
16.
(E)-1-(4-chlorophenyl)-3-(4-(dimethylamino) phenyl) prop-2-en-1-one
A solution of 1-(4-chlorophenyl)ethan-1-one (1.24 g, 8.03 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0 °C. The mixture was stirred for 15 min, after which time 4-(dimethylamino)benzaldehyde (1.0524 g, 7.05 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as an orange solid (1.59 g, 79%). 1H-NMR (300 MHz, CDCl3) δ 3.05 (s, 6H), 6.69 (d, J = 8.9 Hz, 2H), 7.29 (d, J = 15.4 Hz, 1H), 7.45 (d, J = 8.6 Hz, 2H), 7.5 (d, J = 8.9 Hz, 2H), 7.80 (d, J = 15.4 Hz, 1H), 7.95 (d, J = 8.6 Hz, 1H). 13C-NMR (75 MHz, CDCl3) δ 188.74, 163.43, 143.99, 135.07, 130.83, 130.35, 128.93, 128.37, 125.76, 121.86, 113.85, 55.52. Anal. Calcd for C17H16ClNO: C, 71.45; H, 5.64; Cl, 12.41; N, 4.90. Found: C, 71.44; H, 5.68; Cl, 12.21; N, 4.98. Melting point: 137.9–141.9 °C.
17.
(E)-3-(4-(dimethylamino)phenyl)-1-(p-tolyl)prop-2-en-1-one
A solution of 1-(p-tolyl)ethan-1-one (1.37 g, 10.2 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0°C. The mixture was stirred for 15 min, after which time 4-(dimethylamino)benzaldehyde (1.1548 g, 7.74 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as an orange solid (1.44 g, 70%). 1H-NMR (300 MHz, CDCl3) δ 2.42 (s, 3H), 3.04 (s, 6H), 6.77 (d, J = 7.8 Hz, 2H), 7.28 (d, J = 7.8 Hz, 2H), 7.35 (d, J = 15.5 Hz, 1H), 7.55 (d, J = 8.3 Hz, 2H), 7.78 (d, J = 15.5 Hz, 1H), 7.92 (d, J = 7.6 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 190.20, 145.38, 142.89, 136.40, 130.35, 129.17, 128.46, 116.94, 111.87, 40.21, 21.66. Anal. Calcd for C18H19NO: C, 81.47; H, 7.22; N, 5.28. Found: C, 81.70; H, 7.17; N, 5.26. Melting point: 118.6–120.9 °C.
18.
(E)-3-(4-(dimethylamino)phenyl)-1-(3-(trifluoromethyl)phenyl)prop-2-en-1-one
A solution of 1-(3-(trifluoromethyl) phenyl)ethan-1-one (1.08 g, 5.74 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0 °C. The mixture was stirred for 15 min, after which time 4-(dimethylamino)benzaldehyde (1.0043 g, 6.73 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as an orange solid (1.42 g, 66%). 1H-NMR (300 MHz, CDCl3) δ 3.06 (s, 6H), 6.70 (d, J = 8.9 Hz, 2H), 7.30 (d, J = 15.4 Hz, 1H), 7.61 (m, 4H), 7.83 (m, 2H), 8.20 (m, 2H). 13C-NMR (75 MHz, CDCl3) δ 189.05, 152.29, 147.10, 139.67, 131.46, 130.75, 129.08, 128.55, 128.50, 125.12, 125.07, 122.19, 115.75, 111.78, 40.12. Anal. Calcd for C18H16F3NO: C, 67.70; H, 5.05; F, 17.85; N, 4.39. Found: C, 66.94; H, 4.96; F, 17.87; N, 4.22. Melting point: 85.2–86.4 °C.
19.
(E)-1-(4’-bromo-[1,1’-biphenyl]-4-yl)-3-(4-(dimethylamino)phenyl)prop-2-en-1-one
A solution of 1-(4’-bromo-[1,1’-biphenyl]-4-yl)ethan-1-one (1.03 g, 3.74 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0 °C. The mixture was stirred for 15 min, after which time 4-(dimethylamino)benzaldehyde (1.1178 g, 7.49 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as a yellow solid (2.25 g, 74%). 1H-NMR (300 MHz, CDCl3) δ 3.06 (s, 6H), 6.70 (d, J = 7.5 Hz, 2H), 7.36 (d, J = 15.4 Hz, 1H), 7.59 (m, 8H), 7.84 (d, J = 15.3 Hz, 1H), 8.09 (d, J = 6.9 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 189.91, 152.08, 145.97, 143.54, 139.06, 138.12, 132.05, 130.50, 129.02, 128.83, 126.93, 122.59, 122.42, 116.65, 111.82, 40.15. Anal. Calcd for C23H20BrNO: C, 67.99; H, 4.96; Br, 19.67; N, 3.45. Found: C, 67.68; H, 5.05; Br, 19.46; N, 3.39. Melting point: 183.2–187.6 °C.
20.
(E)-3-(4-(dimethylamino)phenyl)-1-phenylprop-2-en-1-one
A solution of acetophenone (1.08 g, 9.01 mmol) in absolute ethanol (10 mL) was added to an aqueous solution of 10% NaOH (30 mL) at 0 °C. The mixture was stirred for 15 min, after which time 4-(dimethylamino)benzaldehyde (0.9784 g, 6.55 mmol) was added. The reaction mixture was then stirred at room temperature for 24 h. The precipitated product was vacuum-filtered and washed with small portions of water/ethanol to yield the desired chalcone as an orange solid (1.33 g, 81%). 1H-NMR (300 MHz, CDCl3) δ 3.05 (s, 6H), 6.70 (d, J = 8.8 Hz, 2H), 7.33 (d, J = 15.5 Hz, 1H) 7.52 (m, 5H), 7.79 (d, J = 15.5 Hz, 1H), 8.00 (d, J = 6.9 Hz, 2H). 13C-NMR (75 MHz, CDCl3) δ 190.72, 152.02, 145.89, 139.08, 139.06, 130.44, 128.47, 128.32, 122.61, 116.87, 111.82, 40.16. Anal. Calcd for C17H17NO: C, 81.24; H, 6.82; N, 5.57. Found: C, 80.81; H, 6.82; N, 5.50. Melting point: 106.9–110.3 °C.
21.
(E)-3-(3-(3-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzonitrile
Boron trifluoride etherate (48% BF3, 781 mg, 5.5 mmol) was added to a stirred solution of 3′-hydroxyacetophenone (150 mg, 1.1 mmol) and 3-cyanobenzaldehyde (289 mg, 2.2 mmol) in 1,4-dioxane (10 mL), and the reaction mixture was heated at 80 °C for 14–24 h. After cooling, the resultant solution was partitioned with EtOAc, washed with 10% HCl (aq), distilled water, and brine, dried over anhydrous Na2SO4, and concentrated in vacuo. The residue was purified using column chromatography (n-hexane:EtOAc = 3:1, 1:1) to obtain 21 as a solid (114 mg, 41%). 1H-NMR (400 MHz, DMSO-d6) δ 9.84 (s, 1H), 8.50 (s, 1H), 8.19 (d, J = 7.8 Hz, 1H), 8.04 (d, J = 15.8 Hz, 1H), 7.90 (dt, J = 7.8, 1.3 Hz, 1H), 7.73 (dt, J = 15.7 Hz, 1H), 7.70–7.63 (m, 2H), 7.51–7.48 (m, 1H), 7.39 (t, J = 7.9 Hz, 1H), 7.08 (ddd, J = 8.1, 2.6, 1.0 Hz, 1H).
22.
(E)-3-(3-(4-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzonitrile
The procedure applied to the synthesis of 21 was used with boron trifluoride etherate (48% BF3, 781 mg, 5.5 mmol), 4-hydroxy acetophenone (150 mg, 1.1 mmol) and 3-cyanobenzaldehyde (289 mg, 2.2 mmol) to obtain 22 as a yellow solid (92 mg, 34%) (Scheme 4). 1H-NMR (400 MHz, DMSO-d6) δ 10.48 (s, 1H), 8.48 (s, 1H), 8.17 (d, J = 8.0 Hz, 1H), 8.13–8.06 (m, 3H), 7.88 (dt, J = 8.0, 1.6 Hz, 1H), 7.69 (d, J = 15.6 Hz, 1H), 7.66 (t, J = 8.0 Hz, 1H), 6.93–6.89 (m, 2H).
23.
(23a) 4-Acetyl-N,N-dipropylbenzenesulfonamide
A mixture of 4-acetylbenzenesulfonyl chloride (300 mg, 1.37 mmol), dipropylamine (151 mg, 1.50 mmol), and triethylamine (277 mg, 2.74 mmol) in anhydrous THF (10 mL) was stirred at room temperature overnight. Water was added, and the reaction mixture was extracted with EtOAc (3 times). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The removal of solvent in vacuo presented as yellow oil (154 mg, 40%).
(23) (E)-4-(3-(3-cyanophenyl)acryloyl)-N,N-dipropylbenzenesulfonamide
The procedure applied to the synthesis of 21 was used with boron trifluoride etherate (48% BF3, 325 mg, 2.29 mmol), 23a (130 mg, 0.46 mmol), and 3-cyanobenzaldehyde (120 mg, 0.92 mmol) to obtain 23 as an ivory fluffy solid (81 mg, 45%) after purification by column chromatography (n-hexane:EtOAc = 10:1, 5:1). 1H-NMR (400 MHz, DMSO-d6) δ 8.52 (s, 1H), 8.35 (d, J = 8.6 Hz, 2H), 8.22 (d, J = 8.0 Hz, 1H), 8.13 (d, J = 15.7 Hz, 1H), 7.99 (d, J = 8.5 Hz, 2H), 7.92 (d, J = 7.8 Hz, 1H), 7.82 (d, J = 15.7 Hz, 1H), 7.69 (t, J = 7.7 Hz, 1H), 3.09 (d, J = 7.6 Hz, 4H), 1.49 (sextet, J = 7.6 Hz, 4H), 0.82 (t, J = 7.6 Hz, 6H).
24.
(24a) 4-Acetyl-N,N-dipentylbenzenesulfonamide
The procedure applied to the synthesis of 23a was used with 4-acetylbenzenesulfonyl chloride (300 mg, 1.37 mmol), diamylamine (235 mg, 1.50 mmol), and triethylamine (277 mg, 2.74 mmol) to obtain 24a as yellow oil (187 mg, 40%).
(24) (E)-4-(3-(3-cyanophenyl)acryloyl)-N,N-dipentylbenzenesulfonamide
The procedure applied to the synthesis of 21 was used with boron trifluoride etherate (48% BF3, 271 mg, 1.91 mmol), 24a (130 mg, 0.38 mmol), and 3-cyanobenzaldehyde (100 mg, 0.77 mmol) to obtain 24 as an off-white crystal (37 mg, 21%) after purification by column chromatography (n-hexane:EtOAc = 10:1). 1H-NMR (400 MHz, DMSO-d6) δ 8.51 (s, 1H), 8.35 (d, J = 8.6 Hz, 2H), 8.22 (d, J = 7.6 Hz, 1H), 8.13 (d, J = 15.7 Hz, 1H), 7.97 (d, J = 8.8 Hz, 2H), 7.92 (dt, J = 7.7, 1.2 Hz, 1H), 7.81 (d, J = 15.7 Hz, 1H), 7.69 (t, J = 7.7 Hz, 1H), 3.11 (t, J = 7.6 Hz, 4H), 1.46 (quintet, J = 7.6 Hz, 4H), 1.29–1.18 (m, 8 H), 0.84 (t, J = 7.2 Hz, 6H).
25.
(2E,4E)-1-(4-methoxyphenyl)-5-phenylpenta-2,4-dien-1-one
4′-methoxyacetophenone (200 mg, 1.33 mmol) and trans-cinnamaldehyde (0.168 mL, 1.33 mmol) were dissolved in absolute ethanol (10 mL) at room temperature. To this stirring solution, 6M NaOH (1 mL) was added dropwise. Precipitate formed instantaneously and the mixture was stirred for 15 min at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a bright yellow powder (321 mg, 91%). 1H-NMR (300 MHz, Chloroform-d) δ 8.00 (d, J = 8.0 Hz, 2H), 7.69–7.53 (m, 1H), 7.49 (d, J = 8.0 Hz, 2H), 7.42–7.27 (m, 3H), 7.11 (d, J = 14.5 Hz, 1H), 7.04–6.88 (m, 4H), 3.86 (s, 3H). 13C-NMR (75 MHz, Chloroform-d) δ 188.66, 163.33, 144.04, 141.43, 136.17, 131.06, 130.70, 129.12, 128.84, 127.25, 127.04, 125.20, 113.81, 55.48. Melting point: 73–76 °C.
26.
(2E,4E)-1-(4-chlorophenyl)-5-(4-(dimethylamino)phenyl)penta-2,4-dien-1-one
4′-chloroacetophenone (0.250 mL, 1.92 mmol), and 4-(dimethylamino)cinnamaldehyde (305 mg, 1.75 mmol) were dissolved in absolute ethanol (15 mL) and THF (1 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6M NaOH (1 mL) was added dropwise during this time. Precipitate slowly formed, and the reaction mixture was stirred for 1 h at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as an orange powder (486 mg, 81%). 1H-NMR (300 MHz, Chloroform-d) δ 7.91 (d, J = 8.5 Hz, 2H), 7.63 (dd, J = 14.7, 10.9 Hz, 1H), 7.44 (d, J = 8.5 Hz, 2H), 7.40 (d, J = 8.9 Hz, 3H), 7.04–6.77 (m, 3H), 6.68 (d, J = 8.9 Hz, 2H), 3.02 (s, 6H). 13C-NMR (75 MHz, Chloroform-d) δ 189.07, 151.14, 146.96, 143.74, 138.55, 137.00, 129.70, 129.05, 128.76, 123.96, 122.17, 121.82, 111.96, 40.22. Melting point: 161–164 °C.
27.
(2E,4E)-5-(4-(dimethylamino)phenyl)-1-(p-tolyl)penta-2,4-dien-1-one
4′-methylacetophenone (0.250 mL, 1.88 mmol) and 4-(dimethylamino)cinnamaldehyde (300 mg, 1.71 mmol) were dissolved in absolute ethanol (15 mL) and THF (1 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6M NaOH (1 mL) was added dropwise during this time. Precipitate slowly formed, and the reaction mixture was stirred for 1 h at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a bright red powder (351 mg, 71%). 1H-NMR (300 MHz, Chloroform-d) δ 7.89 (d, J = 8.2 Hz, 2H), 7.63 (dd, J = 14.8, 10.6 Hz, 1H), 7.40 (d, J = 8.9 Hz, 2H), 7.28 (d, J = 8.1 Hz, 2H), 7.00–6.87 (m, 3H), 6.68 (d, J = 8.9 Hz, 2H), 3.02 (s, 6H), 2.42 (s, 3H). 13C-NMR (75 MHz, Chloroform-d) δ 190.06, 151.03, 145.96, 143.01, 142.81, 136.08, 129.18, 128.85, 128.42, 124.22, 122.64, 122.51, 112.00, 40.23, 21.66. Melting point: 158–160 °C.
28.
(2E,4E)-1-(4-chlorophenyl)-5-phenylpenta-2,4-dien-1-one
To a stirring solution of 4′-chloroacetophenone (0.250 mL, 1.92 mmol) and trans-cinnamaldehyde (0.250 mL, 1.99 mmol), in absolute ethanol (10 mL), 6 M NaOH (1 mL) was added dropwise at room temperature. Precipitate formed instantaneously and the reaction mixture was stirred for an additional 15 min at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a yellow green powder (445 mg, 95%). 1H-NMR (300 MHz, Chloroform-d) δ 7.92 (d, J = 8.6 Hz, 2H), 7.69–7.55 (m, 1H), 7.54–7.43 (m, 4H), 7.37 (m, 3H), 7.10–6.91 (m, 3H). 13C-NMR (75 MHz, Chloroform-d) δ 189.09, 145.34, 142.43, 139.06, 136.48, 135.96, 129.79, 129.78, 129.38, 128.89, 127.35, 126.74, 124.74. Melting point: 138–139 °C.
29.
(E)-2-((E)-3-(4-(dimethylamino)phenyl)allylidene)-2,3-dihydro-1H-inden-1-one
1-indanone (200 mg, 1.51 mmol) and 4-(dimethylamino)cinnamaldehyde (240 mg, 1.38 mmol) were dissolved in absolute ethanol (15 mL) and THF (1 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6M NaOH (1 mL) was added dropwise during this time. Precipitate slowly formed, and the reaction mixture was stirred for 1 h at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as an orange powder (327 mg, 82%). 1H-NMR (300 MHz, Chloroform-d) δ 7.87 (d, J = 7.6 Hz, 1H), 7.56 (m, 2H), 7.48–7.35 (m, 4H), 6.99 (d, J = 15.3 Hz, 1H), 6.84 (dd, J = 15.2, 11.4 Hz,10H), 6.68 (d, J = 8.8 Hz, 2H), 3.82 (s, 2H), 3.02 (s, 6H). 13C-NMR (75 MHz, Chloroform-d) δ 193.63, 151.06, 148.85, 143.14, 139.74, 134.96, 133.89, 133.35, 128.93, 127.34, 126.12, 124.43, 123.96, 119.81, 111.99, 40.22, 30.54. Melting point: 168–170 °C.
30.
(E)-6-methoxy-2-((E)-3-(4-methoxyphenyl)allylidene)-3,4-dihydronaphthalen-1(2H)-one
6-methoxy-1-tetralone (250 mg, 1.42 mmol) and 4-methoxycinnamaldehyde (230 mg, 1.42 mmol) were dissolved in absolute ethanol (10 mL) at room temperature and 6M NaOH (1 mL) was added dropwise. Precipitate slowly formed, and the reaction mixture was stirred for 30 min at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a bright yellow powder (248 mg, 55%). 1H-NMR (300 MHz, Chloroform-d) δ 8.09 (d, J = 8.7 Hz, 1H), 7.52 (d, J = 8.6 Hz, 1H), 7.46 (d, J = 8.6 Hz, 2H), 7.10–6.79 (m, 5H), 6.72 (s, 1H), 3.86 (s, 3H), 3.83 (s, 3H), 2.98 (s, 4H). 13C-NMR (75 MHz, Chloroform-d) δ 186.26, 163.28, 160.21, 145.84, 140.28, 135.83, 133.36, 130.52, 129.55, 128.57, 127.45, 121.49, 114.23, 113.15, 112.33, 55.44, 55.36, 29.19, 25.99. Melting point: 139–140 °C.
31.
(2E,4E)-5-(4-chlorophenyl)-1-(4-nitrophenyl)penta-2,4-dien-1-one
4′-nitroacetophenone (161 mg, 0.972 mmol) and 4-chlorocinnamaldehyde (162 mg, 0.972 mmol) were dissolved in absolute ethanol (15 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6 M NaOH (1 mL) was added dropwise during this time. Precipitate formed instantaneously, and the reaction mixture was stirred at room temperature for 15 min. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a yellow green powder (252 mg, 83%). 1H-NMR (300 MHz, Chloroform-d) δ 8.34 (d, J = 8.9 Hz, 2H), 8.10 (d, J = 9.0 Hz, 2H), 7.62 (ddd, J = 14.9, 6.5, 3.9 Hz, 1H), 7.45 (d, J = 8.7 Hz, 2H), 7.36 (d, J = 8.7 Hz, 2H), 7.11–6.97 (m, 3H). 13C-NMR (75 MHz, Chloroform-d) δ 188.77, 149.97, 146.29, 142.93, 141.99, 135.44, 134.22, 129.29, 129.20, 128.60, 126.95, 124.81, 123.87. Melting point: 122–123 °C.
32.
(E)-2-((E)-3-(4-(dimethylamino)phenyl)allylidene)-2,3-dihydro-1H-inden-1-one
1,3-indandione (200 mg, 1.37 mmol) and trans-cinnamaldehyde (0.190 mL, 1.51 mmol) were dissolved in absolute ethanol (15 mL) at room temperature, and 6 M NaOH (1 mL) was added dropwise. The reaction mixture was stirred at room temperature overnight, then cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a yellow powder (90 mg, 25%). 1H-NMR (300 MHz, Chloroform-d) δ 8.45 (dd, J = 15.5, 12.0 Hz, 1H), 8.03–7.91 (m, 2H), 7.84–7.75 (m, 2H), 7.72–7.59 (m, 3H), 7.43 (m, 3H), 7.34 (d, J = 15.5 Hz, 1H). 13C-NMR (75 MHz, Chloroform-d) δ 151.16, 144.68, 142.14, 135.51, 135.15, 135.03, 130.94, 129.05, 128.70, 123.62, 123.14, 122.97. Melting point: 151–152 °C.
33.
(E)-2-((E)-3-(2-nitrophenyl)allylidene)-3,4-dihydronaphthalen-1(2H)-one
1-tetralone (0.210 mL, 1.58 mmol) and 2-nitrocinnamaldehyde (250 mg, 1.41 mmol) were dissolved in absolute ethanol (20 mL) and THF (2 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6 M NaOH (1 mL) was added dropwise during this time. Precipitate formed instantaneously and the reaction mixture was stirred for 15 min at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a yellow powder (260 mg, 60%). 1H-NMR (300 MHz, Chloroform-d) δ 7.86 (d, J = 7.8 Hz, 1H), 7.76 (d, J = 8.1 Hz, 1H), 7.59 (d, J = 7.9 Hz, 1H), 7.44 (t, J = 7.9 Hz, 1H), 7.35–7.21 (m, 4H), 7.15 (t, J = 7.5 Hz, 1H), 7.08 (d, J = 7.6 Hz, 1H), 6.95 (dd, J = 15.1, 11.7 Hz, 1H), 2.83 (s, 6H). 13C-NMR (75 MHz, Chloroform-d) δ 186.93, 147.71, 143.29, 136.61, 134.59, 134.40, 133.25, 133.21, 133.16, 131.87, 128.95, 128.30, 128.22, 127.96, 127.83, 126.90, 124.69, 28.46, 26.02. Melting point: 188–189 °C.
34.
(2E,4E)-5-(4-(dimethylamino)phenyl)-1-phenylpenta-2,4-dien-1-one
Acetophenone (0.200 mL, 1.71 mmol) and 4-(dimethylamino)cinnamaldehyde (275 mg, 1.56 mmol) were dissolved in absolute ethanol (10 mL) and THF (1 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6 M NaOH (1 mL) was added dropwise during this time. Precipitate slowly formed, and the reaction mixture was stirred for 1 h at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a red flaky solid (327 mg, 76%). 1H-NMR (300 MHz, Chloroform-d) δ 7.98 (d, J = 6.8 Hz, 2H), 7.64 (dd, J = 14.8, 10.6 Hz, 1H), 7.58–7.44 (m, 3H), 7.40 (d, J = 8.9 Hz, 2H), 7.04–6.78 (m, 3H), 6.68 (d, J = 8.9 Hz, 2H), 3.01 (s, 6H). 13C-NMR (75 MHz, Chloroform-d) δ 190.55, 151.09, 146.45, 143.18, 138.71, 132.28, 128.94, 128.48, 128.29, 124.13, 122.58, 122.41, 112.00, 40.22. Melting point: 153–154 °C.
35.
(E)-2-((E)-3-(4-(dimethylamino) phenyl)allylidene)-3,4-dihydronaphthalen-1(2H)-one
1-tetralone (0.200 mL, 1.50 mmol) and 4-(dimethylamino)cinnamaldehyde (240 mg, 1.36 mmol) were dissolved in absolute ethanol (15 mL) and THF (1 mL) at 50 °C. The solution was slowly cooled to room temperature, and 6 M NaOH (1 mL) was added dropwise during this time. Precipitate slowly formed, and the reaction mixture was stirred for 1 h at room temperature. A few chips of ice were added, and the reaction mixture was cooled in an ice bath for 15 min. The precipitate was vacuum-filtered and washed with small portions of cold water/ethanol solution to yield the desired chalcone as a red powder (191 mg, 46%). 1H-NMR (300 MHz, Chloroform-d) δ 8.11 (d, J = 7.7 Hz, 1H), 7.60 (dd, J = 8.1, 2.3 Hz, 1H), 7.44 (m, 3H), 7.35 (t, J = 7.5 Hz, 1H), 7.25 (d, J = 6.5 Hz, 1H), 7.05–6.85 (m, 2H), 6.68 (d, J = 8.7 Hz, 3H), 3.02 (s, 6H), 3.00 (s, 4H). 13C-NMR (75 MHz, Chloroform-d) δ 187.25, 150.88, 143.36, 142.10, 137.61, 134.16, 132.68, 131.51, 128.72, 128.06, 127.97, 126.86, 124.77, 119.12, 112.04, 40.26, 28.79, 25.77. Melting point: 145–147 °C softening, 275–285 °C melting.

4.2. In Vitro Assay

To test the anti-aggregation effects of the different compounds, the SensoLyte Thioflavin T β-Amyloid (142) Aggregation kit was purchased, and assays were performed as described in the literature [16,17,18].

4.2.1. Thioflavin T (ThT) Fluorescence Assay

Samples of ThT (final concentration of 200 μMM) and amyloid-beta (142) peptide (final concentration of 35 μMM) were incubated at 37 °C in a black µClear bottom 96-well plate. The ThT fluorescence intensity of each sample was immediately measured every 5 min for 120 min with 440⁄485 nm excitation/emission filters and with 15 s shaking between reads to facilitate aggregation. An inhibitor control contained Aβ42 and an aggregation inhibitor was supplied (either Morin or Phenol Red) at a final concentration of 100 μM. Positive control contained Aβ42 without inhibitor. The vehicle control contained the assay buffer and DMSO, of concentrations that did not exceed 1%. The tested compound wells contained Aβ42 peptide and either the homotaurine, chalcone, or homotaurine/chalcone derivatives at various concentrations. All the wells were brought to 100 µL as a final volume [16,17,18].

4.2.2. In Vitro Cell Viability Assay

Cell Culture and Exposure

SH-SY5Y cells (CRL-2266, ATCC, Manassas, VA) were maintained in Dulbecco’s Modified Eagle’s Medium and Ham’s F-12 Medium (DMEM:F12) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin and incubated at 37 °C, 5% CO2, and 90% humidity. For the MTT assay, cells were subcultured using trypsin-ethylenediaminetetraacetic acid (EDTA) (0.25%) solution into 96-well plates at a density of 2 × 104 cells/well and allowed to adhere for 24 h. Following the removal of growth media, compounds (IC50), Aβ42 at a final concentration of 20 μM, compound + Aβ42, along with positive (500 μM H2O2) and vehicle controls were added to separate wells in triplicate. Cells were then incubated for 48 h.

Failures

We evaluated LDH release [19], CellTiter Glo [20], and MTS assays [21]. These did not produce any useful results.

MTT Assay and Cell Viability

The MTT [22] assay was used as a measure of cell viability with metabolically active cells reducing the MTT reagent into insoluble formazan. Briefly, following the removal of media, 100 μL of 0.5 mg/mL MTT reagent was added to each well, and the plate was incubated at 37 °C, 5% CO2, and 90% humidity for 3 h. At the end of this incubation, the MTT reagent was removed, and 100 μL of DMSO was added to dissolve the insoluble formazan. Following a 1 h incubation with DMSO at 37 °C, 5% CO2, and 90% humidity, the plate was read at 570 nm.

Statistical Analysis

All statistical analyses were performed using GraphPad Prism 9 (GraphPad, LaJolla, CA). Data were compared by one-way ANOVA followed by Tukey’s post hoc test to compare the differences between all treatment groups (p < 0.05 considered significant). The results are expressed as the mean ± S.E.M. of multiple experiments where n represents the number of individual cell passages.

Author Contributions

Conceptualization, R.P. and M.Y.; methodology, J.P.Q., A.Z., P.G., A.J., K.J.R., G.M.L., S.Y., S.A. and S.R.C.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors wish to thank the Massachusetts College of Pharmacy and Health Sciences University for the financial support of this project.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Health Statistics. CDCWONDER Online Database: About Underlying Cause of Death, 1999–2019. Available online: https://wonder.cdc.gov/ucd-icd10.html (accessed on 13 July 2022).
  2. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s Disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef]
  3. Murphy, M.P.; Levine, H. Alzheimer’s Disease and the Amyloid-β Peptide. J. Alzheimer’s Dis. 2010, 19, 311–323. [Google Scholar] [CrossRef]
  4. Querfurth, H.W.; Laferla, F.M. Alzheimer’s Disease. N. Engl. J. Med. 2018, 10, 329–344. [Google Scholar] [CrossRef] [PubMed]
  5. Madeo, J. The Role of Oxidative Stress in Alzheimer’s Disease. J. Alzheimer’s Dis. Park. 2013, 3, 116–121. [Google Scholar] [CrossRef]
  6. Madav, Y.; Wairkar, S.; Prabhakar, B. Recent Therapeutic Strategies Targeting Beta Amyloid and Tauopathies in Alzheimer’s Disease. Brain Res. Bull. 2019, 146, 171–184. [Google Scholar] [CrossRef]
  7. Arai, T.; Ohno, A.; Mori, K.; Kuwata, H.; Mizuno, M.; Imai, K.; Hara, S.; Shibanuma, M.; Kurihara, M.; Miyata, N.; et al. Inhibition of Amyloid Fibril Formation and Cytotoxicity by Caffeic Acid-Conjugated Amyloid-β C-Terminal Peptides. Bioorg. Med. Chem. Lett. 2016, 26, 5468–5471. [Google Scholar] [CrossRef]
  8. Lee, S.; Zemianek, J.; Shea, T.B. Rapid, Reversible Impairment of Synaptic Signaling in Cultured Cortical Neurons by Exogenously-Applied Amyloid-β. J. Alzheimer’s Dis. 2013, 35, 395–402. [Google Scholar] [CrossRef]
  9. Kumar, D.; Ganeshpurkar, A.; Kumar, D.; Modi, G.; Gupta, S.K.; Singh, S.K. Secretase Inhibitors for the Treatment of Alzheimer’s Disease: Long Road Ahead. Eur. J. Med. Chem. 2018, 148, 436–452. [Google Scholar] [CrossRef]
  10. De La Torre, J.C.; Gonzalez-Lima, F. The FDA Approves Aducanumab for Alzheimer’s Disease, Raising Important Scientific Questions. J. Alzheimer’s Dis. 2021, 82, 881–882. [Google Scholar] [CrossRef] [PubMed]
  11. Erman, W.F.; Kretschmar, H.C. Syntheses and Facile Cleavage of Five-Membered Ring Sultams. J. Org. Chem. 1961, 26, 4841–4850. [Google Scholar] [CrossRef]
  12. Caltagirone, C.; Ferrannini, L.; Marchionni, N.; Nappi, G.; Scapagnini, G.; Trabucchi, M. The Potential Protective Effect of Tramiprosate (Homotaurine) against Alzheimer’s Disease: A Review. Aging Clin. Exp. Res. 2012, 24, 580–587. [Google Scholar] [CrossRef] [PubMed]
  13. Baum, L.; Lam, C.W.K.; Cheung, S.K.-K.; Kwok, T.; Lui, V.; Tsoh, J.; Lam, L.; Leung, V.; Hui, E.; Ng, C.; et al. Six-Month Randomized, Placebo-Controlled, Double-Blind, Pilot Clinical Trial of Curcumin in Patients with Alzheimer Disease. J. Clin. Psychopharmacol. 2008, 28, 110–113. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin Inhibits Formation of Amyloid β Oligomers and Fibrils, Binds Plaques, and Reduces Amyloid in Vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef]
  15. Yang, S.; Shergalis, A.; Lu, D.; Kyani, A.; Liu, Z.; Ljungman, M.; Neamat, M. Design, Synthesis, and Biological Evaluation of Novel Allosteric Protein Disulfide Isomerase Inhibitors. J. Med. Chem. 2019, 62, 3447–3474. [Google Scholar] [CrossRef]
  16. Hellstrand, E.; Boland, B.; Walsh, D.M.; Linse, S. Amyloid β-Protein Aggregation Produces Highly Reproducible Kinetic Data and Occurs by a Two-Phase Process. ACS Chem. Neurosci. 2010, 1, 13–18. [Google Scholar] [CrossRef] [PubMed]
  17. Hudson, S.A.; Ecroyd, H.; Kee, T.W.; Carver, J.A. The Thioflavin T Fluorescence Assay for Amyloid Fibril Detection Can Be Biased by the Presence of Exogenous Compounds. FEBS J. 2009, 276, 5960–5972. [Google Scholar] [CrossRef]
  18. Liu, R.; Barkhordarian, H.; Emadi, S.; Chan, B.P.; Sierks, M.R. Trehalose Differentially Inhibits Aggregation and Neurotoxicity of Beta-Amyloid 40 and 42. Neurobiol. Dis. 2005, 20, 74–81. [Google Scholar] [CrossRef]
  19. Reed, K.J.; Freeman, D.T.; Landry, G.M. Diethylene glycol and its metabolites induce cell death in SH-SY5Y neuronal cells in vitro. Toxicol. In Vitro 2021, 75, 105196. [Google Scholar] [CrossRef]
  20. Dmitriev, R.I.; Papkovsky, D.B. In vitro ischemia decreases histone H4K16 acetylation in neural cells. FEBS Lett. 2015, 589, 138–144. [Google Scholar] [CrossRef]
  21. Kulkarni, N.; Gadde, R.; Gugnani, K.S.; Vu, N.; Yoo, C.; Zaveri, R.; Betharia, S. Neuroprotective effects of disubstituted dithiolethione ACDT against manganese-induced toxicity in SH-SY5Y cells. Neurochem. Int. 2021, 147, 105052. [Google Scholar] [CrossRef]
  22. Martínez, M.-A.; Rodríguez, J.-L.; Lopez-Torres, B.; Martínez, M.; Martínez-Larrañaga, M.-R.; Maximiliano, J.-E.; Anadón, A.; Ares, I. Use of human neuroblastoma SH-SY5Y cells to evaluate glyphosate-induced effects on oxidative stress, neuronal development and cell death signaling pathways. Environ. Int. 2020, 135, 105414. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of homotaurine and curcumin.
Figure 1. Chemical structures of homotaurine and curcumin.
Chemistry 05 00018 g001
Scheme 1. Synthesis of sulfonamide derivative compound (113).
Scheme 1. Synthesis of sulfonamide derivative compound (113).
Chemistry 05 00018 sch001
Figure 2. ThT fluorescence assay. Select sulfonamide derivatives (2, 3, 7) at 100 µM with Phenol red (100 µM) as a positive control. Fluorescence was recorded every 5 min for 120 min.
Figure 2. ThT fluorescence assay. Select sulfonamide derivatives (2, 3, 7) at 100 µM with Phenol red (100 µM) as a positive control. Fluorescence was recorded every 5 min for 120 min.
Chemistry 05 00018 g002
Scheme 2. Synthesis of chalcone derivatives (14–24).
Scheme 2. Synthesis of chalcone derivatives (14–24).
Chemistry 05 00018 sch002
Figure 3. Select ThT fluorescence assay of chalcone compounds 14, 16, and 20 at 100 µM. Phenol red 100 µM is a positive control. Fluorescence was recorded every 5 min for 120 min.
Figure 3. Select ThT fluorescence assay of chalcone compounds 14, 16, and 20 at 100 µM. Phenol red 100 µM is a positive control. Fluorescence was recorded every 5 min for 120 min.
Chemistry 05 00018 g003
Scheme 3. Synthesis of extended chalcone derivatives (25–35).
Scheme 3. Synthesis of extended chalcone derivatives (25–35).
Chemistry 05 00018 sch003
Figure 4. ThT fluorescence assay. Select extended chalcone compounds (25–32) at 100 µM with Phenol red (100 µM) as a positive control. Fluorescence was recorded every 5 min for 120 min.
Figure 4. ThT fluorescence assay. Select extended chalcone compounds (25–32) at 100 µM with Phenol red (100 µM) as a positive control. Fluorescence was recorded every 5 min for 120 min.
Chemistry 05 00018 g004
Figure 5. Amyloid beta alone significantly decreases cell viability in SH-SY5Y cells with compounds 26, 27, and 34 (at IC50 concentrations), providing no significant neuroprotection. Cell viability was measured as an MTT reduction, and data ± SEM (n = 3) were normalized as % vehicle control (black column). Asterisk (*) indicates significant difference from vehicle controls as determined by one-way ANOVA followed by Tukey’s post hoc test (p < 0.05); ns, not significant.
Figure 5. Amyloid beta alone significantly decreases cell viability in SH-SY5Y cells with compounds 26, 27, and 34 (at IC50 concentrations), providing no significant neuroprotection. Cell viability was measured as an MTT reduction, and data ± SEM (n = 3) were normalized as % vehicle control (black column). Asterisk (*) indicates significant difference from vehicle controls as determined by one-way ANOVA followed by Tukey’s post hoc test (p < 0.05); ns, not significant.
Chemistry 05 00018 g005
Scheme 4. Synthesis of 4 substituted acetophenones (23a and 24a).
Scheme 4. Synthesis of 4 substituted acetophenones (23a and 24a).
Chemistry 05 00018 sch004
Table 1. Library and IC50 values of homotaurine derivatives.
Table 1. Library and IC50 values of homotaurine derivatives.
Chemistry 05 00018 i001
Cmpd #IC50RR’R”
1>100 µM Chemistry 05 00018 i002 Chemistry 05 00018 i003H
2>100 µM Chemistry 05 00018 i057 Chemistry 05 00018 i004 Chemistry 05 00018 i005
3>100 µM Chemistry 05 00018 i006 Chemistry 05 00018 i007 Chemistry 05 00018 i008
4>100 µM Chemistry 05 00018 i009 Chemistry 05 00018 i010 Chemistry 05 00018 i011
5>100 µM Chemistry 05 00018 i012 Chemistry 05 00018 i013 Chemistry 05 00018 i014
6>100 µM Chemistry 05 00018 i015 Chemistry 05 00018 i016 Chemistry 05 00018 i017
7>100 µM Chemistry 05 00018 i018 Chemistry 05 00018 i019 Chemistry 05 00018 i020
8>100 µM Chemistry 05 00018 i021 Chemistry 05 00018 i022 Chemistry 05 00018 i023
9>100 µM Chemistry 05 00018 i024 Chemistry 05 00018 i058 Chemistry 05 00018 i025
10>100 µM Chemistry 05 00018 i059 Chemistry 05 00018 i026 Chemistry 05 00018 i027
11>100 µM Chemistry 05 00018 i028 Chemistry 05 00018 i029H
12>100 µM Chemistry 05 00018 i030 Chemistry 05 00018 i031 Chemistry 05 00018 i032
13>100 µM Chemistry 05 00018 i033 Chemistry 05 00018 i034
Table 2. IC50 values of (left) chalcone and chalcone-homotaurine hybrids and (right) extended chalcones. Data reported as triplicates.
Table 2. IC50 values of (left) chalcone and chalcone-homotaurine hybrids and (right) extended chalcones. Data reported as triplicates.
Cmpd #IC50StructureCmpd #IC50Structure
14>100 µM Chemistry 05 00018 i03525>100 µM Chemistry 05 00018 i036
15>100 µM Chemistry 05 00018 i037262.4 µM Chemistry 05 00018 i038
1665.2 µM Chemistry 05 00018 i039273.4 µM Chemistry 05 00018 i040
176.9 µM Chemistry 05 00018 i04128>100 µM Chemistry 05 00018 i042
1833.6 µM Chemistry 05 00018 i0432917.40 µM Chemistry 05 00018 i044
19>100 µM Chemistry 05 00018 i04530>100 µM Chemistry 05 00018 i046
2040.2 µM Chemistry 05 00018 i04731>100 µM Chemistry 05 00018 i048
21>100 µM Chemistry 05 00018 i04932>100 µM Chemistry 05 00018 i050
22>100 µM Chemistry 05 00018 i05133>100 µM Chemistry 05 00018 i052
23>100 µM Chemistry 05 00018 i0533418.68 µM Chemistry 05 00018 i054
24>100 µM Chemistry 05 00018 i0563513.47 µM Chemistry 05 00018 i055
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Quiroz, J.P.; Zeng, A.; Young, M.; Gordon, P.; Jaipuria, A.; Reed, K.J.; Landry, G.M.; Yang, S.; Asher, S.; Zhang, S.R.C.; et al. Homotaurine and Curcumin Analogues as Potential Anti-Amyloidogenic Agents. Chemistry 2023, 5, 223-241. https://doi.org/10.3390/chemistry5010018

AMA Style

Quiroz JP, Zeng A, Young M, Gordon P, Jaipuria A, Reed KJ, Landry GM, Yang S, Asher S, Zhang SRC, et al. Homotaurine and Curcumin Analogues as Potential Anti-Amyloidogenic Agents. Chemistry. 2023; 5(1):223-241. https://doi.org/10.3390/chemistry5010018

Chicago/Turabian Style

Quiroz, Jose Paredes, Andi Zeng, Michelle Young, Patrick Gordon, Aadya Jaipuria, Kristi J. Reed, Greg M. Landry, Suhui Yang, Shreya Asher, Sabrina Ruoyao Chen Zhang, and et al. 2023. "Homotaurine and Curcumin Analogues as Potential Anti-Amyloidogenic Agents" Chemistry 5, no. 1: 223-241. https://doi.org/10.3390/chemistry5010018

Article Metrics

Back to TopTop