Next Article in Journal
Evaluation of Floxuridine Oligonucleotide Conjugates Carrying Potential Enhancers of Cellular Uptake
Previous Article in Journal
Dysregulation of Astrocyte Ion Homeostasis and Its Relevance for Stroke-Induced Brain Damage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis, and Biological Evaluation of Novel 6H-Benzo[c]chromen-6-one Derivatives as Potential Phosphodiesterase II Inhibitors

1
Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China
2
School of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
3
School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou 213164, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(11), 5680; https://doi.org/10.3390/ijms22115680
Submission received: 15 April 2021 / Revised: 21 May 2021 / Accepted: 25 May 2021 / Published: 26 May 2021
(This article belongs to the Section Molecular Biology)

Abstract

:
Urolithins (hydroxylated 6H-benzo[c]chromen-6-ones) are the main bioavailable metabolites of ellagic acid (EA), which was shown to be a cognitive enhancer in the treatment of neurodegenerative diseases. As part of this research, a series of alkoxylated 6H-benzo[c]chromen-6-one derivatives were designed and synthesized. Furthermore, their biological activities were evaluated as potential PDE2 inhibitors, and the alkoxylated 6H-benzo[c]chromen-6-one derivative 1f was found to have the optimal inhibitory potential (IC50: 3.67 ± 0.47 μM). It also exhibited comparable activity in comparison to that of BAY 60-7550 in vitro cell level studies.

Graphical Abstract

1. Introduction

Ellagic acid (EA) is an important part of the pharmacological activity of mellow compounds, which are widely distributed in the bark, root, leaf, and fruit of plants. It has a wide range of pharmacological activities, such as antioxidation, antitumor, antimutation, antibacterial, anti-inflammatory, antiviral, and myocardial protection [1]. In recent years, EA showed great therapeutic potential in the treatment of neurodegenerative diseases such as typical Alzheimer’s disease (AD) and Parkinson’s disease (PD) because of its neuroprotective effects and ability to delay neurodegeneration of aging in the mouse model of AD [2,3,4,5,6,7,8]. However, the applications of EA were limited in the field of health care products and cosmetics due to poor oral bioavailability. There are no EA derivatives available on the market in the pharmaceutical field.
Urolithins are main intestinal metabolites, which are derived from the metabolism of EA through tannolactone ring and dihydroxylation [9,10,11,12]. Urolithins have more advantages in absorption than EA, which may be the final material basis for the biological activities of EA [13]. Urolithins are widely distributed in the urine, feces, and bile of mammals such as humans, rats, mice, cattle, and pigs [13,14,15]. The reports on the biological activity of urolithins mainly focus on the antioxidation, anti-inflammatory, and antitumor properties, as well as its regulation of intestinal flora and inhibition of end products of protein glycosylation [9,10,13,16,17]. Some studies reported that both Urolithin A and Urolithin B are involved in neuroprotection because they can significantly inhibit the formation of protein glycosylation end products [18]. Urolithin A can activate neuroprotective mechanisms, such as activating SIRT-1 in differentiated ReNcell VM human nerve cells. SIRT-1 is a deacetylase that regulates DNA expression, cell apoptosis, and aging, and participates in the physiological or pathological processes of organisms by deacetylating substrate proteins. The activation of SIRT-1 is related to the neuroprotective effect of Urolithin A in nerve cells [19]. Urolithin B can cross the blood-brain barrier and has few side effects on the body, so it may provide therapeutic benefits for neurodegenerative diseases associated with oxidative stress and microglial activation [20]. Therefore, urolithins show great therapeutic potential in the treatment of neurodegenerative diseases.
Phosphodiesterase inhibition received much attention for the potential treatment of central nervous system (CNS) disorders. The role of PDEs in the augmentation of cyclic nucleotide signaling makes these enzymes as attractive targets for the enhancing neuronal communication. PDE2A is the only subtype of PDE2 in mammalian species, which is mainly expressed in brain and heart, but remains low in peripheral tissues [21,22,23,24]. Compared with normal people, the level of PDE2A in the brain of AD patients is increased. Laurent Gomez et al. summarized the development of PDE2 inhibitors for the treatment of cognitive disorders [25,26]. In 2013, Zhu et al. reported the composite crystal structure of PDE2 and the highly selective inhibitor BAY 60-7550 (protein number: 4HTX, http://www.rcsb.org, accessed on 20 March 2021), which provided the basis for the rational design of small molecule ligands targeted to PDE2 [27]. However, there are no PDE2 drugs in the markets so far. Therefore, further studies are urgently needed to develop PDE2 inhibitors to meet clinical needs.
In the present study, urolithins were selected as leading compounds, and a series of novel urolithin(6H-benzo[c]chromen-6-one)-derivatives were designed and synthesized to screen PDE2 inhibitors. The potential neuroprotective effects of resultant compounds were evaluated through PDE2 inhibition effects firstly. The compounds with higher PDE2 inhibition activity were further evaluated by corticosterone-induced cytotoxicity in the hippocampal HT-22 cells.

2. Results and Discussion

2.1. Synthetic Pathways

Our initial exploration focused on introducing different substituents into the hydroxy scaffold at the 3-position of urolithin while keeping the lactone ring fixed. Thus, various kinds of novel 6H-benzo[c]chromen-6-one derivatives were designed and synthesized. The general routes for synthesizing 6H-benzo[c]chromen-6-one derivatives 1 and 2 are shown in Scheme 1 [28]. First, the reaction of 2-bromobenzoic acids and resorcinol in the presence of CuSO4 and NaOH provides the intermediate I and II. Next, intermediate I and II were reacted with various kinds of halides including iodomethane, alkyl bromides, and heterocyclic bromides to generate the desired 6H-benzo[c]chromen-6-one derivatives 1 and 2 in moderate to good yields.

2.2. Enzymatic Assays

Our previous work reported [29] that a propylphenyl group in the classic PDE2 inhibitor BAY 60-7550 was well-accommodated through a hydrophobic-induced binding pocket located under L770 of the protein. Therefore, for urolithin-derivatives in this work, only by bearing certain length and lipophilicity can the R1 group interact properly within the pocket. Based on this rule, the 6H-benzo[c]chromen-6-one derivatives 1 and 2 were evaluated based on their effects on PDE2 inhibitory activities. As shown in Figure 1 and Figure 2, most of the derivatives gave the appropriate values of ClogP (2.0–5.0), indicating the good blood-brain barrier penetration. Moreover, compounds 1a–u and 2a–w were also tested in vitro for PDE2 inhibitory activities, showing that R1 group remarkably affected the PDE2 inhibitory activities (as illustrated in Table 1). For compounds 1a–u, when R1 group alkane substituted groups with less than five carbons, the PDE2 inhibitory activity is relatively good. To our delight, compound 1f significantly affected the PDE2 inhibitory activities (IC50 = 3.67 ± 0.47 μM). For compounds 2aw, although compound 2i had the best PDE2 inhibitory activity, it also needed to be reformed.

2.3. Cell Assays

Corticosterone can induce PDE2A, PDE3B, PDE4A, PDE4D, PDE10, and PDE11 expression in HT-22 cells, which results in significant cell lesion [30,31]. Thus, corticosterone-induced HT-22 neurotoxicity was generally employed as classical cell model to screen PDE-inhibitors for curing neurodegenerative disease, such as AD and PD.
To determine whether compound 1f could promote neuron proliferation and protect HT-22 cells against corticosterone-induced neurotoxicity by PDE2 inhibition, the viability of HT-22 cells was measured in the presence of 100 μM corticosterone (positive control), corticosterone absence (control), and different concentrations of 1f. As shown in Figure 3, the cell viability was significantly decreased when HT-22 cells were exposed to 100 μM corticosterone (p < 0.001), which indicated that HT-22 neurotoxicity model was successfully built. Compound 1f significantly increased cell viability of HT-22 cells in a dose-dependent manner at concentration of 6.25~25 μM, and the best concentration was 12.5 μM (p < 0.01). In addition, the effect was similar with the BAY 60-7550 at a concentration of 1.0 μM. Our results showed compound 1f possessed good protective effects on HT-22 cells, which was associated with PDE2 inhibition.

3. Experiments

3.1. Synthesis

All chemicals, reagents, and solvents were analytical grade, purchased from commercial suppliers (i.e., Aladdin, Adamas, and Acros).
The reactions were monitored via thin-layer chromatography (TLC) performed on HSGF254 plates and visualized under UV light. Melting points were measured with an X4-A microscopic melting point apparatus. 1HNMR spectra were recorded on a BrukerBioSpin GmbH spectrometer at 300 and 400 MHz; 13CNMR spectra were recorded on a BrukerBioSpin GmbH spectrometer at 75 and 100 MHz; the coupling constants are given in Hz. Mass (HRMS) analysis was obtained using Agilent 6200 Accurate-Mass TOF LC/MS system with Electrospray Ionization (ESI). HPLC analysis was obtained using SHIMADZULC-20AB.

3.1.1. 3-Hydroxy-6H-benzo[c]chromen-6-one (I)

2-Bromobenzoic acid (30 mmol, 6.03 g), resorcinol (60 mmol, 6.60 g), sodium hydroxide (60 mmol, 0.8 g), and water (30 mL) were added in the 100 mL sealed tub, and the reaction was stirred 20 min at 100 °C. Then, 5% copper sulfate solution (9.6 mL) was added to the system and heated to reflux for 1 h. The progress of reaction was monitored through TLC (PE:EA = 5:1). At the end of the reaction, the reaction solution was added dropwise to the ice-water mixture, and the filtered solid continued to be washed with water. The filter cake was recrystallized twice with methanol and glacial acetic acid to obtain compound I (3.50 g, 55.0%) [26]. M.p. 250–251 °C. 1H NMR (300 MHz, DMSO-d6) δ = 10.36 (s, 1H), 8.27 (dt, J = 8.0, 3.0 Hz, 1H), 8.21–8.15 (m, 2H), 7.89 (m, 1H), 7.57 (m, 1H), 6.85 (dd, J = 9.0, 3.0 Hz, 1H), 6.76 (d, J = 3.0 Hz, 1H); 13C NMR (75 MHz, DMSO-d6) δ = 161.06, 160.32, 152.57, 135.65, 135.52, 130.11, 128.04, 125.21, 122.02, 119.39, 113.60, 109.82, 103.40; HRMS (ESI) m/z calcd for C13H9O3+[M + H]+: 213.0473; found: 213.0470. Chromatographic purity: 98.0% (HPLC).

3.1.2. General Procedures for Synthesis of Compounds 1au [28]

The anhydrous DMF (30 mL) was added to a 250 mL round bottom flask, compound I (9.4 mmol, 2.0 g), anhydrous K2CO3 (12.2 mmol, 1.7 g), and halide (12.2 mmol) were added, and the temperature was controlled at 80~120 °C; the reaction was stirred for 24 h. The reaction was monitored by TLC (PE:EA = 3:1). When the reaction was over, the reaction solution was added to the ice-water mixture to obtain brown crude solid. After suction filtration and drying, the brown crude solid was purified by column chromatography to obtain compounds 1au.
3-Methoxy-6H-benzo[c]chromen-6-one (1a): Using iodomethane as the starting material, the desired white solid 1a was isolated (1.30 g, 61%). M.p. 131.5–132.8 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.35 (d, J = 8.0 Hz, 1H), 8.29 (d, J = 12.0 Hz, 1H), 8.22 (dd, J = 8.0 Hz, 1H), 7.95–7.90 (m, 1H), 7.62 (t, J = 6.0 Hz, 1H), 7.06–6.99 (m, 2H), 3.88 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 161.67, 160.96, 152.54, 135.78, 135.19, 130.14, 128.53, 125.17, 122.40, 119.68, 112.78, 111.09, 101.91, 56.23; HRMS (ESI) m/z calcd for C14H11O3+[M + H]+: 227.0630; found: 227.0626. Chromatographic purity: 98.2% (HPLC).
Ethoxy-6H-benzo[c]chromen-6-one (1b): Using bromoethane as the starting material, the desired yellow solid 1b was isolated (1.70 g, 75%). M.p. 115.2–117.7 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.35–8.22 (m, 3H), 7.93 (t, J = 6.0 Hz, 1H), 7.62 (t, J = 6.0 Hz, 1H), 6.98 (d, J = 6.0 Hz, 2H), 4.13 (q, J = 3.0 Hz, 2H), 1.39 (t, J = 6.0 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.97, 152.55, 135.81, 135.25, 130.15, 128.52, 125.20, 122.41, 119.68, 113.10, 110.99, 102.31, 64.28, 14.94; HRMS (ESI) m/z calcd for C15H13O3+[M + H]+: 241.0786; found: 241.0782. Chromatographic purity: 97.6% (HPLC).
3-Isopropoxy-6H-benzo[c]chromen-6-one (1c): Using 2-bromopropane as the starting material, the desired yellow solid 1c was isolated (1.70 g, 71%). M.p. 58.7–59.1 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.68–7.33 (m, 5H), 6.96 (s, 2H), 4.77 (m, 1H), 1.32 (d, J = 6.0 Hz, 6H); 13C NMR (75 MHz, DMSO-d6) δ = 160.98, 159.92, 152.59, 135.76, 135.23, 130.13, 128.45, 125.22, 122.34, 119.65, 113.83, 110.83, 103.23, 70.42, 22.13; HRMS (ESI) m/z calcd for C16H15O3+[M + H]+: 255.0943; found: 255.0940. Chromatographic purity: 97.8% (HPLC).
3-Propoxy-6H-benzo[c]chromen-6-one (1d): Using 1-bromopropane as the starting material, the desired white solid 1d was isolated (1.99 g, 83%). M.p. 67.6–69.2 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.28 (t, J = 6.0 Hz, 1H), 8.23–8.17 (m, 2H), 7.92–7.86 (m, 1H), 7.61–7.55 (m, 1H), 6.99–6.93 (m, 2H), 4.04–3.99 (m,2H), 1.83–1.71 (m, 2H), 1.01 (t, J = 6.0 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 161.07, 160.95, 152.50, 135.74, 135.21, 130.11, 128.46,125.12, 122.35,119.64, 113.06, 110.95,102.29, 70.00, 22.35, 10.80; HRMS (ESI) m/z calcd for C16H15O3+[M + H]+: 255.0943; found: 255.0940. Chromatographic purity: 98.9% (HPLC).
3-(Sec-butoxy)-6H-benzo[c]chromen-6-one (1e): Using 2-bromobutane as the starting material, the desired yellow solid 1e was isolated (1.79 g, 71%). M.p. 73.2–75.8 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.36–8.19 (m, 3H), 7.93–7.88 (m, 1H), 7.62–7.57 (m, 1H), 7.00–6.97 (m, 2H), 4.60–4.53 (m, 1H), 1.74–1.58 (m, 2H), 1.28 (d, J = 8.0 Hz, 3H), 0.95 (t, J = 4.0 Hz, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 160.97, 160.25, 152.58, 135.73, 135.24, 130.12, 128.41, 125.19, 122.31, 119.64, 113.83, 110.83, 103.21, 75.23, 28.89, 19.35, 9.92; HRMS (ESI) m/z calcd for C17H17O3+[M + H]+: 269.1099; found: 269.1095. Chromatographic purity: 99.2% (HPLC).
3-butoxy-6H-benzo[c]chromen-6-one (1f): Using 1-bromobutane as the starting material, the desired yellow solid 1f was isolated (1.41 g, 56%). M.p. 53.6–55.5 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.34–8.20 (m, 3H), 7.91 (t, J = 9.0 Hz, 1H), 7.60 (t, J = 9.0 Hz, 1H), 7.01–6.99 (d, J = 6.0 Hz, 2H), 4.09 (t, J = 6.0 Hz, 2H), 1.74(t, J = 9.0 Hz, 2H), 1.50–1.42 (m, 2H), 0.95 (t, J = 9.0 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 161.13, 160.98, 152.54, 135.78, 135.25, 130.14, 128.49, 125.16, 122.39, 119.67, 113.13, 110.98, 102.34, 68.30, 31.03, 19.17, 14.15; HRMS (ESI) m/z calcd for C17H17O3+[M + H]+: 269.1099; found: 269.1095. Chromatographic purity: 98.7% (HPLC).
3-(pentyloxy)-6H-benzo[c]chromen-6-one (1g): Using 1-bromopentane as the starting material, the desired yellow solid 1g was isolated (1.49 g, 56%). M.p. 69.4–71.8 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.34–8.20 (m, 3H), 7.91 (t, J = 8.0 Hz, 1H), 7.60 (t, J = 8.0 Hz, 1H), 7.01–6.98 (m, 2H), 4.08 (t, J = 6.0 Hz, 2H), 1.79–1.72 (m, 2H), 1.46–1.32 (m, 4H), 0.91 (t, J = 8.0 Hz, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 161.10, 160.97, 153.65, 135.77, 135.23, 130.13, 128.47, 125.14, 122.37, 119.66, 113.10, 110.95, 103.11, 68.56, 29.42, 28.12, 22.36, 17.18; HRMS (ESI) m/z calcd for C18H19O3+[M + H]+: 283.1256; found: 283.1252. Chromatographic purity: 99.2% (HPLC).
ethyl 2-((6-oxo-6H-benzo[c]chromen-3-yl)oxy)acetate (1h): Using ethyl 2-bromoacetate as the starting material, the desired yellow solid 1h was isolated (1.40 g, 50%). M.p. 124.7–125.8 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.36–8.21 (m, 3H), 7.93 (t, J = 7.5 Hz, 1H), 7.62 (t, J = 7.5 Hz, 1H), 7.04 (dt, J = 9.0, 3.0 Hz, 2H), 4.94 (s, 2H), 4.20 (q, J = 7.0 Hz, 2H), 1.24 (t, J = 7.5Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 168.79, 160.88, 159.94, 152.37, 136.32, 135.03, 130.17, 128.74, 125.29, 122.52, 120.20, 113.06, 111.79, 103.34, 66.14, 62.31, 15.96; HRMS (ESI) m/z calcd for C17H15O5+[M + H]+: 299.0841; found: 299.0837. Chromatographic purity: 98.3% (HPLC).
3-((tetrahydro-2H-pyran-4-yl)methoxy)-6H-benzo[c]chromen-6-one (1i): Using 4-(bromomethyl)tetrahydro-2H-pyran as the starting material, the desired yellow solid 1i was isolated (1.60 g, 55%). M.p. 133.0–134.8 °C. 1H NMR (300 MHz, CDCl3) δ = 8.34 (d, J = 9.0 Hz, 1H), 7.96 (dd, J = 18.0, 9.0 Hz, 2H), 7.78 (t, J = 7.5 Hz, 1H), 7.50 (t, J = 7.5 Hz, 1H), 6.91–6.82 (m, 2H), 4.04 (dd, J = 12.0, 9.0 Hz, 2H), 3.87 (t, J = 6.0Hz, 2H), 3.46 (t, J = 12.0 Hz, 2H), 2.17–2.06 (m, 1H), 1.80 (d, J = 3.0 Hz, 2H), 1.57–4.13 (m, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 161.09, 160.97, 152.53, 135.79, 135.22, 130.15, 128.53, 125.19, 122.41, 119.69, 113.13, 111.08, 102.44, 73.00, 67.06, 34.77, 29.60; HRMS (ESI) m/z calcd for C19H19O4+[M + H]+: 311.1205; found: 311.1201. Chromatographic purity: 97.3% (HPLC).
3-(Benzyloxy)-6H-benzo[c]chromen-6-one (1j): Using (bromomethyl)benzene as the starting material, the desired yellow solid 1j was isolated (2.81 g, 99%). M.p. 113.2–115.3 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.31 (dd, J = 15.0, 9.0 Hz, 2H), 8.21 (dd, J = 9.0, 1.2 Hz, 1H), 7.94–7.89 (m, 1H), 7.61–7.58 (m, 1H), 7.52–7.48 (m, 2H), 7.45–7.35 (m, 3H), 7.12–7.07 (m, 2H), 5.24 (s, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.95, 160.70, 152.48, 136.92, 135.81, 135.17, 130.16, 128.99, 128.61, 128.53, 128.37, 125.26, 122.47, 119.76, 113.44, 111.34, 102.91, 70.25; HRMS (ESI) m/z calcd for C20H15O3+ [M + H]+: 303.0943; found: 303.0940. Chromatographic purity: 96.8% (HPLC).
3-(2-(1H-pyrazol-1-yl)ethoxy)-6H-benzo[c]chromen-6-one (1k): Using 1-(2-bromoethyl)-1H-pyrazole as the starting material, the desired white solid 1k was isolated (1.24 g, 43%). M.p. 164.3–166.6 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.32 (d, J = 9.0 Hz, 1H), 8.27–8.19 (m, 2H), 7.93–7.88 (m, 1H), 7.82 (d, J = 2.1 Hz, 1H), 7.63–7.57 (m, 1H), 7.48 (d, J = 3.0 Hz, 1H), 7.01–6.95 (m, 2H), 6.27 (t, J = 3.0 Hz, 1H), 4.55 (m, 2H), 4.46 (m, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.94, 160.43, 152.48, 139.39, 135.84, 135.12, 131.08, 130.17, 128.67, 125.29, 122.49, 119.77, 113.16, 111.46, 105.72, 102.63, 67.60, 50.93; HRMS (ESI) m/z calcd for C18H15N2O3+[M + H]+: 307.1004; found: 307.1001. Chromatographic purity: 96.5% (HPLC).
3-(Pyrimidin-2-yloxy)-6H-benzo[c]chromen-6-one (1l): Using 2-bromopyrimidine as the starting material, the desired yellow solid 1l was isolated (1.83 g, 67%). M.p. 174.0–176.6 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.70 (d, J = 8.0 Hz, 2H), 8.45(d, J = 8.0 Hz, 2H), 8.28 (d, J = 12.0 Hz, 1H), 7.98 (t, J = 10.0 Hz, 1H), 7.70 (t, J = 10.0 Hz, 1H), 7.41–7.29 (m, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 164.80, 160.66, 154.69, 151.92, 135.92, 134.55, 130.26, 129.58, 125.31, 123.09, 120.57, 119.02, 117.93, 115.50, 111.00; HRMS (ESI) m/z calcd for C17H11N2O3+[M + H]+: 291.0691; found: 291.0686. Chromatographic purity: 98.2% (HPLC).
3-(2-Hydroxyethoxy)-6H-benzo[c]chromen-6-one (1m): Using 2-bromoethanol as the starting material, the desired white solid 1m was isolated (2.34 g, 98%). M.p. 125.7–126.4 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.32 (d, J = 8.0 Hz, 1H), 8.27–8.24 (m, 1H), 8.21 (dd, J = 8.0, 4.0 Hz, 1H), 7.94–7.90 (m, 1H), 7.61 (t, J = 8.0 Hz, 1H), 7.03–7.00 (m, 2H), 5.01 (t, J = 6.0 Hz, 1H), 4.11 (t, J = 4.0 Hz, 2H), 3.76 (q, J = 4.0 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ = 161.16, 161.07, 152.52, 135.90, 135.23, 130.19, 128.61, 125.23, 122.42, 119.67, 113.25, 111.05, 102.43, 70.65, 59.87; HRMS (ESI) m/z calcd for C15H13O4+[M + H]+: 257.0736; found: 257.0732. Chromatographic purity: 98.3% (HPLC).
3-((Tetrahydrofuran-2-yl)methoxy)-6H-benzo[c]chromen-6-one (1n): Using 2-(bromomethyl)tetrahydrofuran as the starting material, the desired yellow solid 1n was isolated (1.31 g, 47%). M.p. 82.8–84.1 °C. 1H NMR (400 MHz, CDCl3) δ = 8.36 (d, J = 8.0 Hz, 1H), 8.00 (d, J = 8.0 Hz, 1H), 7.94 (d, J = 8.0 Hz, 1H), 7.79 (t, J = 8.0 Hz, 1H), 7.51 (t, J = 8.0 Hz, 1H), 6.96 (dd, J = 8.0, 4.0 Hz, 1H), 6.89 (d, J = 4.0 Hz, 1H), 4.37–4.30 (m, 1H), 4.08–3.95 (m, 3H), 3.91–3.85 (q, J = 8.0 Hz, 1H), 2.18–2.09 (m, 1H), 2.06–1.94 (m, 2H), 1.85–1.76 (m, 1H); 13C NMR (100 MHz, CDCl3) δ = 161.55, 160.74, 152.49, 135.15, 134.88, 130.54, 127.76, 123.77, 121.11, 119.97, 112.90, 111.28, 102.27, 76.85, 70.86, 68.72, 28.19, 25.76; HRMS (ESI) m/z calcd for C18H17O4+[M + H]+: 297.1049; found: 297.1045. Chromatographic purity: 99.1% (HPLC).
3-((1,3-Dioxolan-2-yl)methoxy)-6H-benzo[c]chromen-6-one (1o): Using 2-(bromomethyl)-1,3-dioxolane as the starting material, the desired white solid 1o was isolated 1.93 g, 69%). M.p. 135.6–137.8 °C. 1H NMR (400 MHz, CDCl3) δ = 8.35 (d, J = 8.0 Hz, 1H), 8.00 (d, J = 8.0 Hz, 1H), 7.94 (d, J = 8.0 Hz, 1H), 7.81–7.77 (m, 1H), 7.51 (t, J = 8.0 Hz, 1H), 6.96 (dd, J = 12.0, 4.0 Hz, 1H), 6.90 (d, J = 4.0 Hz, 1H), 5.34 (t, J = 4.0 Hz, 1H), 4.14–3.96 (m, 6H); 13C NMR (100 MHz, CDCl3) δ = 161.45, 160.29, 152.43, 135.02, 134.90, 130.54, 127.86, 123.84, 121.14, 120.01, 112.74, 111.61, 102.49, 101.66, 69.01, 65.41; HRMS (ESI) m/z calcd for C17H15O5+[M + H]+: 299.0841; found: 299.0839. Chromatographic purity: 98.6% (HPLC).
3-((3,5-Dimethoxybenzyl)oxy)-6H-benzo[c]chromen-6-one (1p): Using 1-(bromomethyl)-3,5-dimethoxybenzene as the starting material, the desired white solid 1p was isolated (3.37 g, 99%). M.p. 133.8–135.0 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.23–8.11 (m, 3H), 7.82 (t, J = 9.0 Hz, 1H), 7.52 (t, J = 7.5 Hz, 1H), 7.00– 6.97 (m, 2H), 6.58 (d, J = 3.0 Hz, 2H), 6.40 (t, J = 3.0 Hz, 1H), 5.09 (s, 2H), 3.68 (s, 6H); 13C NMR (75 MHz, DMSO-d6) δ = 161.03, 160.94, 160.56, 152.43, 139.24, 135.76, 135.13, 130.13, 128.58, 125.22, 122.43, 119.73, 113.39, 111.34, 106.01, 102.87, 99.98, 70.04, 55.65; HRMS (ESI) m/z calcd for C22H19O5+[M + H]+: 363.1154; found: 363.1151. Chromatographic purity: 98.4% (HPLC).
3-((1,3-Dimethyl-1H-pyrazol-5-yl)methoxy)-6H-benzo[c]chromen-6-one (1q): Using 5-(bromomethyl)-1,3-dimethyl-1H-pyrazole as the starting material, the desired whitesolid 1q was isolated (2.92 g, 97%). M.p. 165.1–167.6 °C. 1H NMR (300 MHz, CDCl3) δ = 8.34 (dd, J = 9.0, 1.5 Hz, 1H), 8.00–7.93 (m, 2H), 7.82–7.76 (m, 1H), 7.54–7.49 (m, 1H), 6.95 (m, 2H), 6.16 (s, 1H), 5.07 (s, 2H), 3.87 (s, 3H), 2.26 (s, 3H); 13C NMR (75 MHz, CDCl3) δ = 161.39, 159.70, 152.41, 147.46, 136.99, 134.98, 134.89, 130.57, 128.02, 124.00, 121.17, 120.01, 112.96, 111.84, 107.14, 102.50, 60.81, 36.53, 13.44; HRMS (ESI) m/z calcd for C19H17N2O3+[M + H]+: 321.1161; found: 321.1158. Chromatographic purity: 97.1% (HPLC).
3-(Pyridin-3-ylmethoxy)-6H-benzo[c]chromen-6-one (1r): Using 3-(bromomethyl)pyridine as the starting material, the desired yellow solid 1r was isolated (1.42 g, 50%). M.p. 143.6–145.3 °C. 1H NMR (300 MHz, CDCl3) δ = 8.69 (d, J = 30.0 Hz, 2H), 8.35 (dd, J = 9.0, 3.0 Hz, 1H), 7.98 (dd, J = 12.0,6.0 Hz, 2H), 7.853–7.76 (m, 2H), 7.51 (t, J = 7.5 Hz, 1H), 7.37 (dd, J = 9.0, 3.0 Hz, 1H), 6.98 (dd, J = 9.0, 3.0 Hz, 1H), 6.97 (d, J = 3.0 Hz,1H), 5.15 (s, 2H); 13C NMR (75 MHz, CDCl3) δ = 161.39, 160.06, 152.50, 149.79, 149.07, 135.42, 134.95, 134.10, 131.74, 130.58, 127.97, 124.00, 123.65, 121.17, 120.03, 112.92, 111.75, 102.64, 67.94; HRMS (ESI) m/z calcd for C19H14NO3+[M + H]+: 304.0895; found: 304.0891. Chromatographic purity: 96.2% (HPLC).
3-(Cyclopentylmethoxy)-6H-benzo[c]chromen-6-one (1s): Using (bromomethyl)cyclopentane as the starting material, the desired yellow solid 1s was isolated (2.13 g, 77%). M.p. 90.6–91.2 °C. 1H NMR (300 MHz, CDCl3) δ = 8.34 (dd, J = 9.0, 1.5 Hz, 1H), 7.98 (dd, J = 6.0, 1.2 Hz, 1H), 7.91 (d, J = 9.0 Hz, 1H), 7.79–7.74 (m, 1H), 7.51–7.45 (m, 1H), 6.90 (dd, J = 9.0, 3.0 Hz, 1H), 6.83 (d, J = 3.0 Hz, 1H), 3.89 (d, J = 6.0 Hz, 2H), 2.47 (m, 1H), 1.96–1.79 (m, 2H), 1.752–1.55 (m, 4H), 1.44–1.32 (m, 2H); 13C NMR (75 MHz, CDCl3) δ = 161.61, 161.19, 152.56, 135.25, 134.85, 130.52, 127.61, 123.67, 121.05, 119.89, 112.87, 110.87, 102.12, 72.67, 38.90, 29.48, 25.45; HRMS (ESI) m/z calcd for C19H19O3+[M + H]+: 295.1256; found: 295.1251. Chromatographic purity: 98.4% (HPLC).
3-((4-[Hydroxymethyl]benzyl)oxy)-6H-benzo[c]chromen-6-one (1t): Using (4-(bromomethyl)phenyl)methanol as the starting material, the desired white solid 1t was isolated (2.40 g, 77%). M.p. 147.0–149.2 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.33 (d, J = 9.0 Hz, 1H), 8.29–8.21 (m, 2H), 7.96–7.90 (m, 1H), 7.65–7.60 (m, 1H), 7.49 (d, J = 9.0 Hz, 2H), 7.40 (d, J = 9.0 Hz, 2H), 7.12–7.07 (m, 2H), 5.29 (t, J = 9.0 Hz, 1H), 5.24 (s, 2H), 4.55 (d, J = 6.0 Hz, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.95, 160.68, 152.45, 142.94, 135.79, 135.17, 130.15, 128.58, 128.24, 127.02, 125.21, 122.44, 119.72, 113.45, 111.27, 102.87, 70.14, 63.10; HRMS (ESI) m/z calcd for C21H17O4+[M + H]+: 333.1049; found: 333.1044.Chromatographic purity: 97.2% (HPLC).
3-(2-Morpholinoethoxy)-6H-benzo[c]chromen-6-one (1u): Using 4-(2-bromoethyl)morpholine as the starting material, the desired yellow solid 1u was isolated (1.22 g, 40%). M.p. 100.8–102.8 °C. 1H NMR (300 MHz, CDCl3) δ = 8.35 (dd, J = 9.0, 1.2 Hz, 1H), 7.96 (dd, J = 18.0, 6.0 Hz, 2H), 7.78 (m, 1H), 7.50 (t, J = 7.5 Hz, 1H), 6.94–6.90 (dd, J = 9.0, 3.0 Hz, 2H), 4.18 (t, J = 6.0 Hz, 2H), 3.76 (t, J = 7.5 Hz, 4H), 2.85 (t, J = 4.5 Hz, 2H), 2.61 (t, J = 4.5 Hz, 4H); 13C NMR (75 MHz, CDCl3) δ = 161.50, 160.55, 152.52, 135.09, 134.91, 130.55, 127.80, 123.80, 121.10, 119.96, 112.88, 111.29, 102.25, 66.91, 66.25, 57.43, 54.13; HRMS (ESI) m/z calcd for C19H20NO4+[M + H]+: 326.1314; found: 326.1310. Chromatographic purity: 99.2% (HPLC).

3.1.3. General Procedure for the Synthesis of 3-Hydroxy-6H-benzo[c]chromen-6-one (II)

Using 2-bromo-5-methoxybenzoic acid (60 mmol, 13.79 g) acid as the starting material, the synthesis method was similar to that of compound I, the desired yellow solid II was isolated (6.82 g, 47.0%). M.p. 258.4–261.7 °C. 1H NMR (300 MHz, DMSO-d6) δ = 10.22 (s, 1H), 8.21 (d, J = 9.0 Hz, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 3.0 Hz, 1H), 7.49 (dd, J = 9.0, 3.0 Hz, 1H), 6.83 (dd, J = 9.0, 3.0 Hz, 1H), 6.75 (d, J = 3.0 Hz, 1H), 3.89 (s, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.97, 159.41, 158.94, 151.62, 128.97, 124.57, 124.36, 123.99, 120.48, 113.55, 111.27, 109.96, 103.30, 56.01; HRMS (ESI) m/z calcd for C14H10O4+[M+H]+: 242.0579; found: 242.0575. Chromatographic purity: 97.2% (HPLC).

3.1.4. General Procedures for Synthesis of Compounds 2aw

The anhydrous DMF (30 mL) was added to a 250 mL round bottom flask, compound II (9.4 mmol, 2.28 g), anhydrous K2CO3 (12.2 mmol, 1.7 g) and halides (12.2 mmol) were added, and the temperature was controlled at 80~120 °C, the reaction was stirred for 24 h. The reaction was monitored by TLC (PE:EA = 3:1). When the reaction was over, the reaction solution was to the ice-water mixture to obtain brown crude solid. After suction filtration and drying, the brown crude solid was purified by column chromatography to obtain compounds 2aw.
3,8-Dimethoxy-6H-benzo[c]chromen-6-one (2a): Using iodomethane as the starting material, the desired white solid 2a was isolated (1.23 g, 51%). M.p. 146.2–148.8 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.22 (d, J = 9.0 Hz, 1H), 8.14 (d, J = 9.0 Hz, 1H), 7.58 (d, J = 3.0 Hz, 1H), 7.47 (dd, J = 9.0, 3.0 Hz, 1H), 6.96 (s, 1H), 6.94 (t, J = 4.0 Hz, 1H), 3.89 (s, 3H), 3.84 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 160.85, 160.83, 159.23, 151.55, 128.55, 12.50, 124.34, 124.31, 120.79, 112.69, 111.30, 111.21, 101.79, 56.15, 56.04; HRMS (ESI) m/z calcd for C15H13O4+[M + H]+: 257.0736; found: 257.0731. Chromatographic purity: 99.2% (HPLC).
3-Ethoxy-8-methoxy-6H-benzo[c]chromen-6-one (2b): Using bromoethane as the starting material, the desired white solid 2b was isolated(1.14 g, 45%). M.p. 141.1–143.8 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.26 (d, J = 8.0 Hz, 1H), 8.17 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 4.0 Hz, 1H), 7.51 (dd, J = 8.0, 4.0 Hz, 1H), 6.98 (s, 1H), 6.96 (t, J = 4.0 Hz, 1H), 4.13 (q, J = 8.0 Hz, 2H), 3.90 (s, 3H), 1.37 (t, J = 8.0 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.89, 160.10, 159.23, 151.55, 128.61, 124.52, 124.39, 124.32, 120.78, 113.03, 111.32, 111.10, 102.20, 64.19, 56.05, 14.95; HRMS (ESI) m/z calcd for C16H15O4+[M + H]+: 271.0892; found: 271.0890. Chromatographic purity: 98.1% (HPLC).
8-Methoxy-3-propoxy-6H-benzo[c]chromen-6-one (2c): Using 1-bromopropane as the starting material, the desired white solid 2c was isolated (1.39 g, 52%). M.p. 109.3–111.6 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.18 (d, J = 8.0 Hz, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 4.0 Hz, 1H), 7.45 (dd, J = 8.0, 4.0 Hz, 1H), 6.94–6.90 (m, 2H), 3.99 (t, J = 6.0 Hz, 2H), 3.88 (s, 3H), 1.75 (m, 2H), 1.00 (t, J = 8.0 Hz, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 160.86, 160.23, 159.20, 151.52, 128.58, 124.46, 124.33, 124.27, 120.75, 113.00, 111.29, 111.08, 102.20, 69.95, 56.02, 22.37, 10.81; HRMS (ESI) m/z calcd for C17H17O4+[M + H]+: 285.1049; found: 285.1045. Chromatographic purity: 99.3% (HPLC).
3-(Sec-butoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2d): Using 2-bromobutane as the starting material, the desired yellow solid 2d was isolated (0.95 g, 34%). M.p. 64.5–66.7 °C. 1H NMR (400 MHz, DMSO-d6) δ = 8.21 (d, J = 8.0 Hz, 1H), 8.12 (d, J = 8.0 Hz, 1H), 7.59 (d, J = 4.0 Hz, 1H), 7.47 (dd, J = 8.0, 4.0 Hz, 1H), 6.95 (s, 1H), 6.92 (d, J = 4.0 Hz, 1H), 4.56–4.49 (m, 1H), 3.89 (s, 3H), 1.73–1.57 (m, 2H), 1.27 (d, J = 6.0 Hz, 3H), 0.94 (t, J = 8.0 Hz, 3H); 13C NMR (100 MHz, DMSO-d6) δ = 160.89, 159.40, 159.20, 151.62, 128.62, 124.57, 124.35, 124.27, 120.77, 113.84, 111.32, 111.01, 103.23, 75.18, 56.03, 28.90, 19.37, 9.94; HRMS (ESI) m/z calcd for C18H19O4+[M + H]+: 299.1205; found: 299.1201. Chromatographic purity: 98.7% (HPLC).
3-Butoxy-8-methoxy-6H-benzo[c]chromen-6-one (2e): Using 1-bromobutane as the starting material, the desired white solid 2e was isolated (1.82 g, 65%). M.p. 102.5–104.4 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.26 (d, J = 9.0 Hz, 1H), 8.19–8.15 (m, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.51 (dd, J = 9.0, 3.0 Hz, 1H), 6.98 (s, 1H), 6.96 (t, J = 4.5 Hz, 1H), 4.07 (t, J = 6.0 Hz, 2H), 3.90 (s, 3H), 1.78–1.68 (m, 2H), 1.52–1.3840 (m, 2H), 0.95 (t, J = 7.5 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.84, 160.25, 159.20, 151.52, 128.58, 124.43, 124.29, 124.24, 120.75, 112.99, 111.31, 111.08, 102.21, 68.21, 56.02, 31.06, 19.17, 14.14; HRMS (ESI) m/z calcd for C18H19O4+[M + H]+: 299.1205; found: 299.1201. Chromatographic purity: 99.1% (HPLC).
8-Methoxy-3-(pentyloxy)-6H-benzo[c]chromen-6-one (2f): Using 1-bromopentane as the starting material, the desired white solid 2f was isolated (1.34 g, 46%). M.p. 73.1–75.5 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.23 (d, J = 9.0 Hz, 1H), 8.15–8.12 (m, 1H), 7.59 (d, J = 3.0 Hz, 1H), 7.49 (dd, J = 8.0, 3.0 Hz, 1H), 6.97–6.93 (m, 2H), 4.03 (t, J = 7.5 Hz, 2H), 3.89 (s, 3H), 1.78–1.69 (m, 2H), 1.44–1.34 (m, 4H), 0.93–0.88 (m, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.89, 160.26, 159.22, 151.54, 128.62, 124.50, 124.39, 124.32, 120.77, 113.05, 111.29, 111.09, 102.21, 68.49, 56.04, 28.70, 28.13, 22.37, 14.40; HRMS (ESI) m/z calcd for C19H21O4+[M + H]+: 313.1362; found: 313.1359. Chromatographic purity: 98.5% (HPLC).
3-Isopropoxy-8-methoxy-6H-benzo[c]chromen-6-one (2g): Using 2-bromopropane as the starting material, the desired white solid 2g was isolated (0.97 g, 36%). M.p. 88.3–90.7 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.21 (d, J = 9.0 Hz, 1H), 8.14–8.11 (m, 1H), 7.58 (d, J = 3.0 Hz, 1H), 7.48 (dd, J = 9.0, 3.0 Hz, 1H), 6.95–6.91 (m, 2H), 4.75 (m, 1H), 3.89 (s, 3H), 1.32 (d, J = 6.0 Hz, 6H); 13C NMR (75 MHz, DMSO-d6) δ = 160.89, 159.19, 159.05, 151.60, 128.60, 124.55, 124.34, 124.26, 120.76, 113.81, 111.30, 110.98, 103.20, 70.33, 56.03, 22.14; HRMS (ESI) m/z calcd for C17H17O4+[M + H]+: 285.1049; found: 285.1045. Chromatographic purity: 98.6% (HPLC).
3-(2-Hydroxyethoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2h): Using 2-bromoethanol as the starting material, the desired yellow solid 2h was isolated (1.51 g, 56%). M.p. 166.8–168.0 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.21 (d, J = 9.0 Hz, 1H), 8.15–8.11 (m, 1H), 7.57 (d, J = 3.0 Hz, 1H), 7.47 (dd, J = 9.0, 3.0 Hz, 1H), 6.98–6.94 (m, 2H), 4.98 (t, J = 6.0 Hz, 1H), 4.08 (dd, J = 6.0, 3.0 Hz, 2H), 3.89 (s, 3H), 3.77 (q, J = 6.0 Hz, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.86, 160.25, 159.19, 151.50, 128.55, 124.48, 124.33, 124.26, 120.76, 113.05, 111.26, 111.14, 102.28, 70.56, 59.89, 56.02; HRMS (ESI) m/z calcd for C16H15O5+[M + H]+: 287.0841; found: 287.0837. Chromatographic purity: 98.8% (HPLC).
8-Methoxy-3-(2-(methylthio)ethoxy)-6H-benzo[c]chromen-6-one (2i): Using (2-bromoethyl)(methyl)sulfane as the starting material, the desired white solid 2i was isolated (1.18 g, 40%). M.p. 110.1–111.8 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.27 (d, J = 9 Hz, 1H), 8.19 (d, J = 9.0 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.52 (dd, J = 9.0, 3.0 Hz, 1H), 7.03–6.98 (m, 2H), 4.26 (t, J = 6.0 Hz, 2H), 3.90 (s, 3H), 2.89 (t, J = 7.5 Hz, 2H), 2.18 (s, 3H); 13C NMR (75 MHz, CDCl3) δ = 165.60, 164.53, 164.05, 156.28, 133.29, 129.33, 129.13, 125.61, 117.84, 116.16, 116.12, 107.21, 72.70, 60.82, 37.28, 20.45; HRMS (ESI) m/z calcd for C17H17O4S+[M + H]+: 317.0769; found: 317.0765. Chromatographic purity: 97.8% (HPLC).
Ethyl 2-((8-methoxy-6-oxo-6H-benzo[c]chromen-3-yl)oxy)acetate (2j): Using ethyl 2-bromoacetate as the starting material, the desired white solid 2j was isolated (1.58 g, 51%). M.p. 151.5–152.8 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.28 (d, J = 9.0 Hz, 1H), 8.21 (t, J = 6.0 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.52 (dd, J = 9.0, 3.0 Hz, 1H), 7.03– 6.99 (m, 2H), 4.92 (s, 2H), 4.20 (q, J = 6.0 Hz, 2H), 3.90 (s, 3H), 1.23 (t, J = 7.5 Hz, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 168.85, 160.80, 159.42, 159.11, 151.39, 128.39, 124.63, 124.48, 124.40, 120.98, 113.01, 111.93, 111.42, 102.75, 65.41, 61.25, 56.09, 14.52; HRMS (ESI) m/z calcd for C18H17O6+[M + H]+: 329.0947; found: 329.0943. Chromatographic purity: 97.4% (HPLC).
3-(Cyclopentylmethoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2k): Using (bromomethyl)cyclopentane as the starting material, the desired white solid 2k was isolated (0.90 g, 30%). M.p. 146.1–148.2 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.26 (d, J = 9.0 Hz, 1H), 8.17 (t, J = 4.5 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.51 (dd, J = 9.0, 3.0 Hz, 1H), 6.99–6.95 (m, 2H), 3.94 (d, J = 6.0 Hz, 2H), 3.90 (s, 3H), 2.38–2.28 (m, 1H), 1.82–1.74 (m, 2H), 1.65–1.51 (m, 4H), 1.38–1.27 (m, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.92, 160.44, 159.28, 151.59, 128.66, 124.55, 124.44, 124.39, 120.83, 113.15, 111.39, 111.15, 102.35, 72.61, 56.09, 38.88, 29.44, 25.41; HRMS (ESI) m/z calcd for C20H21O4+[M + H]+: 325.1362; found: 325.1360. Chromatographic purity: 98.1% (HPLC).
8-Methoxy-3-((tetrahydrofuran-2-yl)methoxy)-6H-benzo[c]chromen-6-one (2l): Using 2-(bromomethyl)tetrahydrofuran as the starting material, the desired white solid 2l was isolated (1.71 g, 56%). M.p. 113.1–115.5 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.27 (d, J = 9.0 Hz, 1H), 8.18 (dd, J = 9.0, 3.0 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.51 (dd, J = 9.0, 3.0 Hz, 1H), 7.03–6.94 (m, 2H), 4.23–4.15 (m, 1H), 4.11–3.98 (m, 2H), 3.90 (s, 3H), 3.84–3.77 (m, 1H), 3.73–3.66 (m, 1H), 2.08–1.99 (m, 1H), 1.94–1.77 (m, 2H), 1.72–1.65 (m, 1H); 13C NMR (75 MHz, DMSO-d6) δ = 160.86, 160.14, 159.27, 151.51, 128.57, 124.52, 124.36, 120.83, 113.09, 111.35, 111.29, 102.39, 76.81, 71.08, 67.97, 56.06, 28.06, 25.67; HRMS (ESI) m/z calcd for C19H19O5+[M + H]+: 327.1154; found: 327.1150. Chromatographic purity: 99.2% (HPLC).
3-((1,3-Dioxolan-2-yl)methoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2m): Using 2-(bromomethyl)-1,3-dioxolane as the starting material, the desired white solid 2m was isolated (0.93 g, 30%). M.p. 147.9–150.1 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.28 (d, J = 9.0 Hz, 1H), 8.19 (d, J = 9.0 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.52 (dd, J = 9.0, 3.0 Hz, 1H), 7.04–6.99 (m, 2H), 5.24 (t, J = 3.0 Hz, 1H), 4.12 (d, J = 6.0 Hz, 2H), 4.01–3.96 (m, 2H), 3.90 (s, 3H), 3.89– 3.85 (m, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.87, 159.76, 159.38, 151.49, 128.52, 124.63, 124.48, 124.45, 120.94, 113.12, 111.62, 111.41, 102.62, 101.57, 69.02, 65.03, 56.10; HRMS (ESI) m/z calcd for C18H17O6+[M + H]+: 329.0947; found:329.0943. Chromatographic purity: 97.6% (HPLC).
3-(Cyclohexylmethoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2n): Using (bromomethyl)cyclohexane as the starting material, the desired white solid 2n was isolated (1.85 g, 38%). M.p. 139.8–140.1 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.25 (d, J = 9.0 Hz, 1H), 8.18–8.14 (m, 1H), 7.61 (d, J = 3.0 Hz, 1H), 7.50 (dd, J = 9.0, 3.0 Hz, 1H), 6.99–6.95 (m, 2H), 3.90 (s, 3H), 3.87 (d, J = 6.3 Hz, 2H), 1.84–1.65 (m, 6H), 1.33–1.04 (m, 5H); 13C NMR (75 MHz, DMSO-d6) δ = 160.91, 160.43, 159.26, 151.58, 128.66, 124.54, 124.42, 124.37, 120.81, 113.10, 111.37, 111.12, 102.33, 73.67, 56.08, 37.43, 29.65, 26.49, 25.70; HRMS (ESI) m/z calcd for C21H23O4+[M + H]+: 339.1518; found: 339.1514. Chromatographic purity: 97.5% (HPLC).
3-(2-(1,3-Dioxolan-2-yl)ethoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2o): Using 2-(2-bromoethyl)-1,3-dioxolane as the starting material, the desired white solid 2o was isolated (1.22 g, 38%). M.p. 151.6–153.2 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.27 (d, J = 9.0 Hz, 1H), 8.20–8.17 (m, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.51 (dd, J = 9.0, 3.0 Hz, 1H), 7.00–6.96 (m, 2H), 5.02 (t, J = 4.5 Hz, 1H), 4.18 (t, J = 6.0 Hz, 2H), 3.95–3.91 (m, 2H), 3.90 (s, 3H), 3.83–3.79 (m, 2H), 2.07 (m, 2H); 13C NMR (75 MHz, CDCl3) δ = 161.64, 159.88, 159.13, 151.57, 128.66, 124.48, 123.11, 122.82, 120.96, 112.70, 111.33, 110.92, 102.20, 101.86, 65.02, 64.08, 55.76, 33.59, 29.73; HRMS (ESI) m/z calcd for C19H19O6+[M + H]+: 343.1103; found: 343.1101. Chromatographic purity: 96.9% (HPLC).
8-Methoxy-3-((tetrahydro-2H-pyran-4-yl)methoxy)-6H-benzo[c]chromen-6-one (2p): Using 4-(bromomethyl)tetrahydro-2H-pyran as the starting material, the desired yellow solid 2p was isolated (1.79 g, 36%). M.p. 168.1–168.5 °C. 1H NMR (400 MHz, CDCl3) δ = 7.94 (d, J = 8.0 Hz, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 4.0 Hz, 1H), 7.39 (dd, J = 8.0, 4.0 Hz, 1H), 6.91 (dd, J = 8.0, 4.0 Hz, 1H), 6.86 (d, J = 4.0 Hz, 1H), 4.06 (dd, J = 12.0, 4.0 Hz, 2H), 3.94 (s, 3H), 3.88 (d, J = 8.0 Hz, 2H), 3.51–3.45 (m, 2H), 2.19–2.0 (m, 1H), 1.82–1.78 (m, 2H), 1.56–1.46 (m, 2H); 13C NMR (75 MHz, DMSO-d6) δ = 160.91, 160.29, 159.31, 151.59, 128.64, 124.60, 124.45, 124.41, 120.86, 113.14, 111.41, 111.25, 102.42, 72.99, 67.06, 56.10, 34.79, 29.62; HRMS (ESI) m/z calcd for C20H21O5+[M + H]+: 341.1311; found: 341.1307. Chromatographic purity: 97.4% (HPLC).
3-(Benzyloxy)-8-methoxy-6H-benzo[c]chromen-6-one (2q): Using (bromomethyl)benzene as the starting material, the desired white solid 2q was isolated (1.04 g, 33%). M.p. 141.7–142.5 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.27 (d, J = 9.0 Hz, 1H), 8.20 (d, J = 9.0 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.51–7.48 (m, 3H), 7.45–7.33 (m, 3H), 7.10–7.04 (m, 2H), 5.22 (s, 2H), 3.90 (s, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 162.53, 159.77, 158.99, 151.43, 136.98, 128.96, 128.33, 124.30, 121.58, 120.60, 113.25, 111.25, 102.70, 70.15, 55.98; HRMS (ESI) m/z calcd for C21H17O4+[M + H]+: 333.1049; found: 333.1045. Chromatographic purity: 98.7% (HPLC).
3-((4-[Hydroxymethyl]benzyl)oxy)-8-methoxy-6H-benzo[c]chromen-6-one (2r): Using (4-(bromomethyl)phenyl)methanol as the starting material, the desired white solid 2r was isolated (1.26 g, 37%). M.p. 192.4–194.6 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.27 (d, J = 9.0 Hz, 1H), 8.19 (d, J = 9.0 Hz, 1H), 7.62 (d, J = 3.0 Hz, 1H), 7.51 (dd, J = 9.0, 3.0 Hz, 1H), 7.44 (dd, J = 6.0, 3.0 Hz, 2H), 7.35 (d, J = 8.0 Hz, 2H), 7.08–7.03 (m, 2H), 5.20 (t, J = 6.0 Hz, 3H), 4.51 (d, J = 3.0 Hz, 2H), 3.90 (s, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.86, 159.87, 159.32, 151.49, 142.91, 135.27, 128.55, 128.18, 127.01, 124.57, 124.40, 120.88, 113.43, 111.44, 111.39, 102.84, 70.11, 63.11, 56.08; HRMS (ESI) m/z calcd for C22H19O5+[M + H]+: 363.1154; found: 363.1150. Chromatographic purity: 98.6% (HPLC).
3-((3,5-Dimethoxybenzyl)oxy)-8-methoxy-6H-benzo[c]chromen-6-one (2s): Using 1-(bromomethyl)-3,5-dimethoxybenzene as the starting material, the desired white solid 2s was isolated (1.59 g, 43%). M.p. 153.6–155.3 °C. 1H NMR (300 MHz, CDCl3) δ = 7.89 (d, J = 9.0 Hz, 1H), 7.84 (d, J = 9.0 Hz, 1H), 7.73 (d, J = 3.0 Hz, 1H), 7.35 (dd, J = 9.0, 3.0 Hz, 1H), 6.96 (dd, J = 9.0, 3.0 Hz, 1H), 6.89 (d, J = 3.0 Hz, 1H), 6.59 (d, J = 3.0 Hz, 2H), 6.42 (t, J = 3.0 Hz, 1H), 5.05 (s, 2H), 3.91 (s, 3H), 3.80 (s, 6H); 13C NMR (75 MHz, DMSO-d6) δ = 161.04, 160.88, 160.82, 159.76, 159.33, 151.48, 139.36, 128.54, 124.62, 124.44, 120.90, 113.41, 111.51, 111.35, 106.65, 105.99, 102.85, 99.96, 70.00, 56.08, 55.67, 55.49; HRMS (ESI) m/z calcd for C23H21O6+[M + H]+: 393.1260; found: 393.1257. Chromatographic purity: 98.3% (HPLC).
3-((2-Fluorobenzyl)oxy)-8-methoxy-6H-benzo[c]chromen-6-one (2t):Using 1-(bromomethyl)-2-fluorobenzene as the starting material, the desired white solid 2t was isolated (2.47 g, 75%). M.p. 203.6–204.9 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.30 (d, J = 9.0 Hz, 1H), 8.23 (d, J = 9.0 Hz, 1H), 7.64–7.58 (m, 2H), 7.52 (dd, J = 9.0, 3.0 Hz, 1H), 7.49–7.42 (m, 1H), 7.33–7.24 (m, 2H), 7.15 (d, J = 3.0 Hz, 1H), 7.07 (dd, J = 9.0, 3.0 Hz, 1H), 5.25 (s, 2H), 3.91 (s, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 159.49, 159.27, 151.59, 130.17, 130.06, 129.80, 128.58, 124.55, 124.42, 124.37, 123.26, 122.89, 121.09, 115.70, 115.42, 112.90, 111.78, 111.00, 102.74, 64.17, 55.79; HRMS (ESI) m/z calcd for C21H16FO4+[M + H]+: 351.0954; found: 351.0950. Chromatographic purity: 97.6% (HPLC).
3-((1,3-Dimethyl-1H-pyrazol-5-yl)methoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2u): Using 5-(bromomethyl)-1,3-dimethyl-1H-pyrazole as the starting material, the desired white solid 2u was isolated (1.71 g, 52%). M.p. 184.6–186.6 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.29 (d, J = 9.0 Hz, 1H), 8.21 (d, J = 9.0 Hz, 1H), 7.63 (d, J = 3.0 Hz, 1H), 7.52 (dd, J = 9.0, 3.0 Hz, 1H), 7.15 (d, J = 3.0 Hz, 1H), 7.07 (dd, J = 9.0, 3.0 Hz, 1H), 6.19 (s, 1H), 5.23 (s, 2H), 3.90 (s, 3H), 3.76 (s, 3H), 2.12 (s, 3H); 13C NMR (75 MHz, CDCl3) δ = 165.59, 164.15, 164.09, 156.20, 150.86, 142.74, 133.24, 129.37, 129.23, 129.16, 125.71, 118.13, 116.53, 116.17, 111.87, 107.73, 65.66, 60.85, 18.37; HRMS (ESI) m/z calcd for C21H19N2O4+[M + H]+: 351.1267; found: 351.1263. Chromatographic purity: 98.7% (HPLC).
8-Methoxy-3-(pyrimidin-2-yloxy)-6H-benzo[c]chromen-6-one (2v): Using 2-bromopyrimidine as the starting material, the desired white solid 2v was isolated (1.29 g, 23%). M.p. 212.3–214.0 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.69 (d, J = 6.0 Hz, 2H), 8.38 (dd, J = 9.0, 6.0 Hz, 2H), 7.68 (d, J = 3.0 Hz, 1H), 7.57 (dd, J = 9.0, 3.0 Hz, 1H), 7.38 (d, J = 3.0 Hz, 1H), 7.33 (t, J = 4.5 Hz, 1H), 7.27 (dd, J = 9.0, 3.0 Hz, 1H), 3.93 (s, 3H); 13C NMR (75 MHz, CDCl3) δ = 160.11, 158.86, 152.24, 149.99, 126.84, 123.51, 122.38, 122.22, 120.83, 117.40, 115.74, 114.80, 110.18, 109.98, 54.79; HRMS (ESI) m/z calcd for C18H13N2O4+[M + H]+: 321.0797; found: 321.0794. Chromatographic purity: 96.8% (HPLC).
3-(2-(1H-pyrazol-1-yl)ethoxy)-8-methoxy-6H-benzo[c]chromen-6-one (2w): Using 1-(2-bromoethyl)-1H-pyrazole as the starting material, the desired white solid 2w was isolated (1.93 g, 61%). M.p. 159.0–161.1 °C. 1H NMR (300 MHz, DMSO-d6) δ = 8.32 (d, J = 9.0 Hz, 1H), 8.22 (d, J = 9.0 Hz, 1H), 7.87 (d, J = 3.0 Hz, 1H), 7.67 (d, J = 3.0 Hz, 1H), 7.58–7.53 (m, 2H), 7.05–6.98 (m, 2H), 6.32 (t, J = 3.0 Hz, 1H), 4.59 (t, J = 6.0 Hz, 2H), 4.50 (t, J = 4.5 Hz, 2H), 3.95 (s, 3H); 13C NMR (75 MHz, DMSO-d6) δ = 160.83, 159.59, 159.35, 151.49, 139.37, 131.06, 128.47, 124.61, 124.41, 120.90, 113.10, 111.60, 111.39, 105.70, 102.55, 67.55, 56.08, 50.95; HRMS (ESI) m/z calcd for C19H17N2O4+[M + H]+: 337.1110; found: 337.1106. Chromatographic purity: 98.9% (HPLC).

3.2. Enzymatic Assays

The in vitro inhibitory activity of compounds 1au and 2aw against PDE2, which used Bio-cAMP as substrate, are listed in Table 1. The specific experimental method was reported below [32].
Preparation of 1*Rection Buffer: we took 1 mL 10*Rection Buffer, added 9 mL ultra-pure water to swirl and mix well, and put it on ice; preparation of PDE2 solution: 210 μL of 1*Rection Buffer were added into a centrifuge tube, then 30 μL PDE2 were added, mixed well, equal to 8-times dilution, and placed on ice; preparation of compound solution with gradient concentration: DMSO was used as the solvent to prepare 10 mM compound solution, and then diluted with 1*Rection Buffer. The solution with final concentrations of 40 nM, 20 nM, 10 nM, 5 nM, 2.5 nM, 1.25 nM, and 0.625 nM was prepared, respectively, and the solution was vortex mixed.
We took 2 μL of the diluted compound solution of each concentration into the microplate, added 4 μL PDE2 solution, numbered them A1–A9, added 2 μL 1*Rection Buffer and 4 μL PDE2 into A10, added 2 μL compound solution and 4 μL 1*Rection Buffer to A11, added 6 μL 1*Rection Buffer to A13, and ensured the three groups are parallel to each other. Centrifugation was carried out at 1000 R and 25 °C for about 2 min. The reaction time was 0.5 h in the microporous plate thermostatic oscillator. After the reaction, 4 μL Bio-cAMP was added to A1–A13, and centrifugation was carried out at 1000 R and 25 °C for about 2 min and reacted in a microporous plate thermoelectric oscillator for 1 h.
Anti-Camp Alohascreen Acceptor and Streptavidin Coated Donor Bead were purchased from Perkin Elmer Company according to response volume: 1* Immunoassay Buffer 549.32 μL and 3.70 μL anti-cAMP AlohaScreen Acceptor were collected under dark conditions. We took 1.98 μL Streptavidin coated Donor Bead, mixed well, added 15 μL mixture to each well after 1 h reaction, and reacted it in the dark for 1 h. After the reaction, we tested it with multilabel microplate detector. According to the detection results, the inhibition rate was calculated according to the dilution ratio of the experimental system and the formula, and the IC50 of compound was calculated by GraphPad Prism 5.

3.3. Cell Assays

3.3.1. Solution Preparation

We added 943 µL of DMSO solution to 8.2 mg of dexamethasone acetate to obtain a 20 mM stock solution, froze it at −20 °C, and diluted it to 2 mM with serum-containing medium before use. The test compound was dissolved in DMSO to obtain a 20 mM stock solution and stored at −20 °C. Diluted with serum-containing medium to a concentration of 2 × 100, 2 × 50, 2 × 25, 2 × 12.5, 2 × 6.25, 2 × 3.125 μM before use. The control sample Bay 60-7550 stock solution with a concentration of 1 mM was diluted with serum-containing medium to a concentration of 2 × 0.1, 2 × 1, 2 × 2, and 2 × 4 μM before use.

3.3.2. Cell Culture and Dexamethasone Intervention

After resuscitation, HT-22 cells were cultured in DMEM medium (containing 10% FBS) at 37 °C with 5% CO2. After the cells grew to more than 80% cell fusion, we passaged and seeded the plate, and the density of each well should be 10,000 cells/100 μL/well and cultured for 24 h. After the cells adhered to the wall, drug intervention was given.

3.3.3. Drug Intervention

10 µL solution of 2 mM dexamethasone acetate was cultured at 37 °C for 30 min. 100 µL drug solution of 2 × 100, 2 × 50, 2 × 25, 2 × 12.5, 2 × 6.25, and 2 × 3.125 μM were added to make the final concentration of dexamethasone acetate 100 μM, and the final concentrations of the drug were 100, 50, 25, 12.5, 6.25, 3.125 μM. After preparation, it took another 24 h to continue the culture. There were 5 replicate wells for each sample, and the normal control group, dexamethasone acetate-affected group, and positive control group were set at the same time (final concentration 0.1, 1, 2, and 4 μM).

3.3.4. MTT Color Experimental and Data Processing

After the incubation, 50 µL of MTT was added to each well, and it needed another 4 h to incubate. Discarded the upper matrix and the 150 μL DMSO was added. To dissolve the blue-violet precipitate completely, the shaker was used to shake for 10 min. Finally, the wavelength A 490 was measured. The data were expressed as mean ± sd (n = 5), and one-way anova analysis of variance was used for data statistics; p < 0.05 is defined as a significant difference.

4. Conclusions

In this paper, a series of new 6H-benzo[c] chromen-6-one derivatives were designed and synthesized with PDE2 as the potential therapeutic target, and the PDE2 inhibitory activity was tested at the enzyme level. The results showed that part of compounds exhibited good PDE2 inhibitory activity, such as 1f, 1h, 1i, 1l, 1s, 2e, and 2i. Among them, 1f has the highest PDE2 inhibitory activity with IC50 value of 3.67 ± 0.47 μM. The protective effects of compound 1f against corticosterone-induced HT-22 neurotoxicity were further verified. Compound 1f can protect HT-22 cells from corticosterone-induced death and rescue corticosterone-induced toxicity in a dose-dependent manner, which is probably associated with PDE2 inhibition. In the future, we will design and synthesize more active PDE2-inhibitors based on an analysis of the structure-activity relationship between compound 1f and 4HTX for the potential treatment of AD. In addition, the in vivo test would be performed in mouse models of AD to obtain more convincing and potentially therapeutic molecules.

Author Contributions

Synthesis, materials preparation and biological studies, L.T., Y.T., Y.X.; Synthesis and characterization, L.T., Y.X.; Biological activity studies and Validation, Y.W., Z.Z.; Supervision, J.J., G.S.; Writing-original draft, L.T., X.F.; Writing—review and editing, L.T., X.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, grant number 81803471.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shakeri, A.; Zirak, M.R.; Sahebkar, A. Ellagic Acid: A logical Lead for Drug Development. Curr. Pharm. Des. 2018, 24, 106–122. [Google Scholar] [CrossRef]
  2. Rojanathammanee, L.; Puig, K.L.; Combs, C.K. Pomegranate Polyphenols and Extract Inhibit Nuclear Factor of Activated T-Cell Activity and Microglial Activation in Vitro and in a Transgenic Mouse Model of Alzheimer Disease. J. Nutr. 2013, 143, 597–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. West, T.; Atzeva, M.; Holtzman, D.M. Pomegranate Polyphenols and Resveratrol Protect the Neonatal Brain Against Hypoxic-Ischemic Injury. Dev. Neurosci. 2007, 29, 363–372. [Google Scholar]
  4. Campos-Esparza, M.D.R.; Torres-Ramos, M.A. Neuroprotection by Natural Polyphenols: Molecular Mechanisms. Cent. Nerv. Syst. Agents Med. Chem. 2010, 10, 269–277. [Google Scholar] [CrossRef] [PubMed]
  5. Hartman, R.E.; Shah, A.; Fagan, A.M.; Schwetye, K.E.; Parsadanian, M.; Schulman, R.N.; Finn, M.B.; Holtzman, D.M. Pomegranate Juice Decreases Amyloid Load and Improves Behavior in a Mouse Model of Alzheimer’s Disease. Neurobiol. Dis. 2006, 24, 506–515. [Google Scholar] [CrossRef] [PubMed]
  6. Choi, S.J.; Lee, J.H.; Heo, H.J.; Cho, H.Y.; Kim, H.K.; Kim, M.O.; Sun, S.H.; Shin, D.H. Punica Granatum Protects against Oxidative Stress in PC12 Cells and Oxidative Stress-Induced Alzheimer’s Symptoms in Mice. J. Med. Food 2011, 14, 695–701. [Google Scholar] [CrossRef]
  7. Kujawska, M.; Jourdes, M.; Kurpik, M.; Szulc, M.; Szaefer, H.; Chmielarz, P.; Kreiner, G.; Kuźniak, V.K.; Mikołajczak, P.L.; Teissedre, P.L.; et al. Neuroprotective Effects of Pomegranate Juice against Parkinson’s Disease and Presence of Ellagitannins-Derived Metabolite—Urolithin A—in the Brain. Int. J. Mol. Sci. 2020, 21, 202. [Google Scholar] [CrossRef] [Green Version]
  8. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the Gut Barrier Integrity by a Microbial Metabolite Through the Nrf2 Pathway. Nat. Commun. 2019, 10, 2369–2379. [Google Scholar]
  9. Garazd, Y.L.; Garazd, M.M. Natural Dibenzo[b,d]pypan-6-ones: Structural Diversity and Biological Activity. Chem. Nat. Compd. 2016, 52, 1–18. [Google Scholar] [CrossRef]
  10. Cozza, G.; Gianoncelli, A.; Bonvini, P.; Zorzi, E.; Pasquale, R.; Rosolen, A.; Pinna, L.A.; Meggio, F.; Zagotto, G.; Moro, S. Urolithin as a Converging Scaffold Linking Ellagic Acid and Coumarin Analogues Design of Potent Protein Kinase CK2 Inhibitors. Chem. Med. Chem. 2011, 6, 2273–2286. [Google Scholar] [CrossRef]
  11. García-Villalba, R.; Selma, M.V.; Espín, J.C.; Tomas-Barberan, F.A. Identification of Novel Urolithin Metabolites in Human Feces and Urine after the Intake of a Pomegranate Extract. J. Agric. Food Chem. 2019, 67, 11099–11107. [Google Scholar] [CrossRef] [Green Version]
  12. Gaya, P.; Peirotén, Á.; Medina, M.; Álvarez, I.; Landete, J.M. Bifidobacterium Pseudocatenulatum INIA P815: The First Bacterium Able to Produce Urolithins A and B from Ellagic Acid. J. Funct. Foods 2018, 45, 95–99. [Google Scholar] [CrossRef]
  13. Espin, J.C.; Larrosa, M.; García-Conesa, M.T.; Tomás-Barberán, F. Biological Significance of Urolithins, the Gut Microbial Ellagic Acid Derived Metabolites: The Evidence So Far. Evid. Based Complementary Altern. 2013, 2013, 270418. [Google Scholar] [CrossRef] [Green Version]
  14. Tulipani, S.; Urpi-Asrda, M.; Garcia-Villalba, R.; Rabassa, M.; Lopez-Uriarte, P.; Bullo, M.; Jauregui, O.; Tomas-Barberan, F.; Salas-Salvado, J.; Espin, J.C.; et al. Urolithins Are the Main Urinary Microbial-Derived Phenolic Metabolites Discriminating A Moderate Consumption of Nuts in Free-Living Subjects with Diagnosed Metabolic Syndrome. J. Agric. Food Chem. 2012, 60, 8930–8940. [Google Scholar] [CrossRef]
  15. Gonzalez-Barrio, R.; Truchado, P.; Garcia-Villaba, R.; Hervas, G.; Frutos, P.; Espin, J.C.; Tomas-Barberan, F. Metabolism of Oak Leaf Ellagitannins snd Urolithin Production in Beef Cattle. J. Agric. Food Chem. 2012, 60, 3068–3077. [Google Scholar] [CrossRef] [PubMed]
  16. Larrosa, M.; Gonzalez-Sarrias, A.; Yanez-Gasscon, M.J.; Selma, M.V.; Azorin-Ortuno, M.; Toti, S.; Tomas-Barberan, F.; Dolara, P.; Espin, J.C. Anti-Inflammatory Properties of a Pomegranate Extract and Its Metabolite Urolithin-A in a Colitis Rat Model and the Effect of Colon Inflammation on Phenolic Metabolism. J. Nutr. Biochem. 2010, 21, 717–725. [Google Scholar] [CrossRef] [PubMed]
  17. Sharma, M.; Li, L.Y.; Celver, J.; Killian, C.; Kovoor, A.; Seearm, N.P. Effects of Fruit Ellagitannin Extracts, Ellagic Acid, and Their Colonic Metabolite, Urolithin A, on Wnt Signaling. J. Agric. Food Chem. 2010, 58, 3965–3969. [Google Scholar] [CrossRef] [Green Version]
  18. Liu, W.X.; Ma, H.; Frost, L.; Yuan, T.; Dain, J.A.; Seeram, N.P. Pomegranate Phenolics Inhibit Formation of Advanced Glycation Endproducts by Scavenging Reactive Carbonyl Species. Food Funct. 2014, 5, 2996–3004. [Google Scholar] [CrossRef] [PubMed]
  19. Velagapudi, R.; Lepiarz, I.; El-Bakoush, A.; Katola, F.O.; Bhatia, H.; Fiebich, B.L.; Olajide, O. Induction of Autophagy and Activation of SIRT-1 Deacetylation Mechanisms Mediate Neuroprotection by the Pomegranate Metabolite Urolithin A in BV2 Microglia and Differentiated 3D Human Neural Progenitor Cells. Mol. Nutr. Food Res. 2019, 63, 1801237. [Google Scholar] [CrossRef] [Green Version]
  20. Lee, G.; Park, J.S.; Lee, E.J.; Ahn, J.H.; Kim, H.S. Anti-Inflammatory and Antioxidant Mechanisms of Urolithinb in Activated Microglia. Phytomedicine 2019, 55, 50–57. [Google Scholar] [CrossRef]
  21. Lyketsos, C.G.; Lopez, O.; Jones, B.; Fitzpatrick, A.L.; Breitner, J.; DeKosky, S. Prevalence of Neuropsychiatric Symptoms in Dementia and Mild Cognitive Impairment: Results from the Cardiovascular Health Study. JAMA J. Am. Med. Assoc. 2002, 288, 1475–1483. [Google Scholar] [CrossRef] [Green Version]
  22. Bender, A.T.; Beavo, J.A. Cyclic Nucleotide Phosphodiesterases: Molecular Regulation to Clinical Use. Pharmacol. Rev. 2006, 58, 488–520. [Google Scholar] [CrossRef]
  23. Beavo, J.A.; Brunton, L.L. Cyclic Nucleotide Research—Stillexpanding After Half a Century. Nat. Rev. Mol. Cell Biol. 2002, 3, 710–718. [Google Scholar] [CrossRef]
  24. Lugnier, C. Cyclic Nucleotide Phosphodiesterase (PDE) Superfamily: A New Target for the Development of Specific Therapeuticagents. Pharmacol. Therapeut. 2006, 109, 366–398. [Google Scholar] [CrossRef] [PubMed]
  25. Jiang, M.Y.; Han, C.; Zhang, C.; Zhou, Q.; Zhang, B.; Le, M.L.; Huang, M.X.; Wu, Y.N.; Luo, H.B. Discovery of Effective Phosphodiesterase 2 Inhibitors with Antioxidant Activities for the Treatment of Alzheimer’s Disease. Bioorg. Med. Chem. Lett. 2021, 41, 128016. [Google Scholar] [CrossRef]
  26. Gomez, L.; Xu, R.; Sinko, W.; Selfridge, B.; Vernier, W.; Ly, K.; Truong, R.; Metz, M.; Marrone, T.; Sebring, K.; et al. Mathematical and Structural Characterization of Strong Nonadditive Structure-Activity Relationship Caused by Protein Conformational Changes. J. Med. Chem. 2018, 61, 7754–7766. [Google Scholar] [CrossRef] [PubMed]
  27. Zhu, J.; Yang, Q.Q.; Dai, D.D.; Huang, Q. X-Ray Crystal Structure of Phosphodiesterase 2 in Complex with a Highly Selective, Nanomolar Inhibitor Reveals a Binding-Induced Pocket Important for Selectivity. J. Am. Chem. Soc. 2013, 135, 11708–11711. [Google Scholar] [CrossRef]
  28. Gulcan, H.O.; Unlu, S.; Esiringu, İ.; Ercetin, T.; Sahin, Y.; Oz, D.; Sahin, M.F. Design, Synthesis and Biological Evaluation of Novel 6H-Benzo[C]Chromen-6-One, and 7,8,9,10-Tetrahydro-Benzo[C]Chromen-6-One Derivatives as Potential Cholinesterase Inhibitors. Bioorg. Med. Chem. 2014, 22, 5141–5154. [Google Scholar] [CrossRef] [PubMed]
  29. Huang, X.F.; Cao, Y.J.; Zhen, J.; Zhang, D.W.; Kong, R.; Jiang, W.T.; Xu, Y.; Song, G.Q.; Ke, H.M.; Liu, L. Design, Synthesis of Novel Purin-6-One Derivatives as Phosphodiesterase 2 (PDE2) Inhibitors: The Neuroprotective and Anxiolytic-Like Effects. Bioorg. Med. Chem. Lett. 2019, 29, 481–486. [Google Scholar] [CrossRef]
  30. Zhu, M.J.; Shi, J.; Chen, Y.; Huang, G.B.; Zhu, X.W.; Zhang, S.; Huang, X.F.; Song, G.Q.; Zhang, H.T.; Ke, H.M.; et al. Phosphodiesterase 2 Inhibitor Hcyb1 Reverses Corticosterone-Induced Neurotoxicity and Depression-Like Behavior. Psychopharmacology 2020, 237, 3215–3224. [Google Scholar] [CrossRef] [PubMed]
  31. Xu, Y.; Pan, J.C.; Chen, L.; Zhang, C.; Sun, J.; Li, J.X.; Nguyen, L.; Nair, N.; Zhang, H.T.; O’Donnell, J. Phosphodiesterase-2 Inhibitor Reverses Corticosterone-Induced Neurotoxicity and Related Behavioural Changes via Cgmp/PKG Dependent Pathway. Int. J. Neuropsychopharmacol. 2013, 16, 835–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Xu, H.; Xu, X.S.; Gao, Y.; Cao, Y.J.; Huang, X.F.; Kong, R.; Song, G.Q. Establishment of a High-Throughput Screening Model for Drugs Targeting Phosphodiesterase 2. Chin. J. New Drugs 2020, 29, 56. [Google Scholar]
Scheme 1. Synthesis of 6H-benzo[c]chromen-6-one derivatives 1 and 2. Reagents and conditions: (a) 5% CuSO4, NaOH, H2O, reflux, 1.2 h; (b) K2CO3, DMF, 80~120 °C, 3.5~30 h.
Scheme 1. Synthesis of 6H-benzo[c]chromen-6-one derivatives 1 and 2. Reagents and conditions: (a) 5% CuSO4, NaOH, H2O, reflux, 1.2 h; (b) K2CO3, DMF, 80~120 °C, 3.5~30 h.
Ijms 22 05680 sch001
Figure 1. CLoP values of 6H-benzo[c]chromen-6-one derivatives 1.
Figure 1. CLoP values of 6H-benzo[c]chromen-6-one derivatives 1.
Ijms 22 05680 g001
Figure 2. CLoP values of 6H-benzo[c]chromen-6-one derivatives 2.
Figure 2. CLoP values of 6H-benzo[c]chromen-6-one derivatives 2.
Ijms 22 05680 g002
Figure 3. Compound 1f protected cells against corticosterone (CORT)-induced cytotoxicity in a concentration-dependent manner. HT-22 cells were treated with 100 M CORT for 30 min and compound 1f was added for 24 h. Cell viability was measured by MTS assay. Results are expressed as the mean ± standard error of the mean (SEM) of six independent experiments performed in triplicates. *** p < 0.001, compared to control group; # p < 0.05 and ## p < 0.01, compared to vehicle-treated CORT group.
Figure 3. Compound 1f protected cells against corticosterone (CORT)-induced cytotoxicity in a concentration-dependent manner. HT-22 cells were treated with 100 M CORT for 30 min and compound 1f was added for 24 h. Cell viability was measured by MTS assay. Results are expressed as the mean ± standard error of the mean (SEM) of six independent experiments performed in triplicates. *** p < 0.001, compared to control group; # p < 0.05 and ## p < 0.01, compared to vehicle-treated CORT group.
Ijms 22 05680 g003
Table 1. In vitro PDE2 inhibitory activity of 6H-benzo[c]chromen-6-one derivatives 1 and 2.
Table 1. In vitro PDE2 inhibitory activity of 6H-benzo[c]chromen-6-one derivatives 1 and 2.
CompoundPDE2 IC50 a (μM)CompoundPDE2 IC50 a,b (μM)
1a28.82 ± 7.82a>50
1b25.40 ± 5.132b>50
1c25.80 ± 5.452c>50
1d>502d>50
1e37.65 ± 4.92e17.77 ± 4.07
1f3.67 ± 0.472f>50
1g>502g30.29 ± 7.38
1h18.69 ± 2.722h>50
1i17.88 ± 1.872i14.88 ± 1.78
1j43.22 ± 3.672j>50
1k>502k>50
1l19.27 ± 2.382l>50
1m>502m>50
1n27.51 ± 1.492n>50
1o>502o>50
1p>502p25.16 ± 6.91
1q>502q23.31 ± 4.28
1r27.56 ± 2.142r>50
1s8.14 ± 0.542s>50
1t>502tInsoluble
1u>502u29.39 ± 5.01
2v23.88 ± 545
2w>50
a BAY 60-7550 was used as the reference compound with an IC50 of 8.4 nM. b Results are expressed as the mean of at least three experiments.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tang, L.; Jiang, J.; Song, G.; Wang, Y.; Zhuang, Z.; Tan, Y.; Xia, Y.; Huang, X.; Feng, X. Design, Synthesis, and Biological Evaluation of Novel 6H-Benzo[c]chromen-6-one Derivatives as Potential Phosphodiesterase II Inhibitors. Int. J. Mol. Sci. 2021, 22, 5680. https://doi.org/10.3390/ijms22115680

AMA Style

Tang L, Jiang J, Song G, Wang Y, Zhuang Z, Tan Y, Xia Y, Huang X, Feng X. Design, Synthesis, and Biological Evaluation of Novel 6H-Benzo[c]chromen-6-one Derivatives as Potential Phosphodiesterase II Inhibitors. International Journal of Molecular Sciences. 2021; 22(11):5680. https://doi.org/10.3390/ijms22115680

Chicago/Turabian Style

Tang, Long, Jianchun Jiang, Guoqiang Song, Yajing Wang, Ziheng Zhuang, Ying Tan, Yan Xia, Xianfeng Huang, and Xiaoqing Feng. 2021. "Design, Synthesis, and Biological Evaluation of Novel 6H-Benzo[c]chromen-6-one Derivatives as Potential Phosphodiesterase II Inhibitors" International Journal of Molecular Sciences 22, no. 11: 5680. https://doi.org/10.3390/ijms22115680

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop