Next Article in Journal
Coumarin-Induced Hepatotoxicity: A Narrative Review
Next Article in Special Issue
2-Phenylpyridine Derivatives: Synthesis and Insecticidal Activity against Mythimna separata, Aphis craccivora, and Tetranychus cinnabarinus
Previous Article in Journal
Screening of the Active Compounds against Neural Oxidative Damage from Ginseng Phloem Using UPLC-Q-Exactive-MS/MS Coupled with the Content-Effect Weighted Method
Previous Article in Special Issue
Recent Advances in DMSO-Based Direct Synthesis of Heterocycles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Characterization and Biological Evaluation of Benzothiazole–Isoquinoline Derivative

1
Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316022, China
2
Pharmacy College, Jilin Medical University, Jilin 132013, China
3
Key Laboratory of Natural Resource of the Changbai Mountain and Functiaonal Molecules, Ministry of Education, Yanbian University, Yanji 133000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(24), 9062; https://doi.org/10.3390/molecules27249062
Submission received: 8 November 2022 / Revised: 2 December 2022 / Accepted: 2 December 2022 / Published: 19 December 2022

Abstract

:
Currently, no suitable clinical drugs are available for patients with neurodegenerative diseases complicated by depression. Based on a fusion technique to create effective multi–target–directed ligands (MTDLs), we synthesized a series of (R)–N–(benzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl) acetamides with substituted benzothiazoles and (S)–1–phenyl–1,2,3,4–tetrahydroisoquinoline. All compounds were tested for their inhibitory potency against monoamine oxidase (MAO) and cholinesterase (ChE) by in vitro enzyme activity assays, and further tested for their specific inhibitory potency against monoamine oxidase B (MAO–B) and butyrylcholinesterase (BuChE). Among them, six compounds (4b4d, 4f, 4g and 4i) displayed excellent activity. The classical antidepressant forced swim test (FST) was used to verify the in vitro results, revealing that six compounds reduced the immobility time significantly, especially compound 4g. The cytotoxicity of the compounds was assessed by the MTT method and Acridine Orange (AO) staining, with cell viability found to be above 90% at effective compound concentrations, and not toxic to L929 cells reversibility, kinetics and molecular docking studies were also performed using compound 4g, which showed the highest MAO–B and BuChE inhibitory activities. The results of these studies showed that compound 4g binds to the primary interaction sites of both enzymes and has good blood–brain barrier (BBB) penetration. This study provides new strategies for future research on neurodegenerative diseases complicated by depression.

1. Introduction

Neurodegenerative diseases represent an important health issue because of the aging population [1]. Alzheimer’s disease (AD) accounts for the largest proportion of people suffering from neurodegenerative disorders, followed by Parkinson’s disease (PD) [2,3,4]. AD is rapidly becoming an economic burden to nations, with mortality rates associated with AD increasing [5,6,7]. In addition, depressive symptoms are prevalent in individuals with neurodegenerative diseases, and according to recent meta–analysis research, depression is present in roughly 40% of AD cases [8], and can be fatal [9].
Notably, depression may be a risk factor associated with AD development, and neurodegeneration in the AD brain may also predispose individuals to depression [10,11]. AD and depression have distinct neuropathological features. Nonetheless, there are some similarities in the mechanisms (e.g., dysregulation of monoaminergic neurotransmitters, decreased nutritional support, HPA axis disruption, neuroinflammation, excitotoxicity and oxidative damage) that may play a role in the neurodegenerative processes caused by two illnesses [12,13,14,15,16,17,18,19,20]. The use of antidepressants in AD is linked with severe side effects such as hyponatremia [21], cardiotoxicity [22] and an increase in possible hemorrhaging [23,24,25]. In one meta–analysis, treatment of neurodegenerative diseases, especially AD comorbid depression included selective serotonin reuptake inhibitors (SSRIs), noradrenergic and specific serotonergic antidepressants, as well as reversible selective monoamine oxidase inhibitors. These drugs have also been studied without significant treatment effects [26,27,28].
The use of antidepressants to treat neurodegenerative diseases has not shown substantial therapeutic benefit, and additional research on the disease pathways and pathophysiology of neurodegenerative diseases complicated by depression is required to elucidate the pathogenesis and identify the preventive treatment of these diseases [29,30,31,32]. Dual–drug or drug–combination techniques and enhancement strategies are likely to be effective in the combined treatment of these diseases. Currently, therapy to treat neurodegenerative diseases for patients with comorbid depression remains an urgent, unmet global issue. Unfortunately, no clinically authorized medicine that prevents or reduces the progression of the illness has been identified.
The goal of this study was to develop compounds that cure symptoms of depression while also preventing neurodegeneration, thereby confirming the validity of our medicinal design concept. In this report, we used a fusion technique to create effective multi–target–directed ligands (MTDLs) [33,34,35] and combined this approach with previous research findings [36,37,38,39], as shown in Figure 1. Benzothiazole (fragment A) is a heterocycle often used in drug discovery research [40,41]. With various benzothiazole–bearing compounds displaying potential therapeutic effects against AD and depression, this pharmacophoric category is essential for drug development against the neurodegenerative disease comorbid depression sector [42,43,44,45]. Sabeluzole, a benzothiazole–based agent, has been demonstrated to slow the clinical course of AD [46]. In addition, isoquinoline (fragment C) has a diverse set of biological activities, including antidepressant, anti–AD, anti–PD and other neuroprotective activities [47,48,49,50,51]. Benzothiazole (fragment A) and isoquinoline (fragment C) are connected by an amide bond (fragment B) because amide compounds are used to treat illnesses of the central nervous system, such as anxiety, schizophrenia, epilepsy and depression, and have antioxidant pharmacological activity [52,53,54,55]. The most relevant parameters of the newly created MTDLs include physicochemical features of the compounds in connection to their drug–likeness and success in the drug development process. The bulk of the compounds have met Lipinski’s rule of five criteria and bioavailability requirements [56]. In conclusion, the designed and synthesized series of (R)–N–(benzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinoline–2(1H)–yl)acetamides have good potential (Figure 1). The compounds can be a potential therapy for neurodegenerative disease and depression comorbidities and as useful reference prototypes for the developing new treatment strategies.

2. Results and Discussion

2.1. Chemistry

Synthetic Route and Method of Novel Benzothiazole–Isoquinoline Derivatives

The synthetic methods used to obtain the target derivatives are presented in Scheme 1. The structures of the new derivatives were verified by HPLC, IR, 1H NMR, 13C NMR and mass spectrometry analyses.
In the IR spectra of 4a4p derivatives, an NH stretching band at 3289–3261 cm−1 for the single bond was observed, and a CO band for the amide double bond (1619–1512 cm−1) appeared, which supports target amidation. In the 1H NMR spectrum, –NH proton signals were observed at 9.15–10.35 ppm. The α–CH2 proton signals of the amides of 4a4p were observed at 3.95–4.79 ppm, and the C2–H protons in 1–phenyl–3,4–dihydro–isoquinolin–2(1H)–yl were observed to resonate at 4.72–6.79 ppm. In addition, the 13C NMR spectra of 4a4p also confirmed the presence of amide bonds, benzothiazole groups and 1–phenyl–3,4–dihydroisoquinolin groups. The experimental section contains additional synthetic pathways and spectroscopic details.

2.2. In Vitro Biological Evaluation

2.2.1. Inhibitory Activity of Derivatives on MAO

Derivatives (4a4p) were tested for MAO inhibitory activity, and pargyline, rasagiline and clorgyline were used as reference compounds. Table 1 displays the results. We discovered that the majority of benzothiazole–isoquinoline derivatives have excellent MAO inhibitory activity. Among them, analog 4g (IC50 = 14.80 ± 5.45 μM) had the strongest antagonistic effect on MAO activity, and analog 4d (IC50 = 64.83 ± 4.20 μM) had the highest inhibitory rate on MAO (43.37%).
The results of the structure–activity relationship (SAR) for the benzothiazole–isoquinoline derivatives on MAO are shown in Table 1. The results for compound 4g (57.11%, IC50 = 14.80 ± 5.45 μM) revealed that introducing an o–CH3 group on the benzothiazole ring increased MAO inhibition. In contrast, a sharp decrease in MAO inhibition was observed for compound 4h (16.72%, IC50 = 76.37 ± 2.58 μM) and 4i (26.46%, IC50 = 18.53 ± 1.69 μM) suggesting that the o–CH3 group is essential for inhibiting activity of MAO. In addition to the o–CH3 group, compounds that included an electron–withdrawing group significantly enhanced MAO antagonism. Moreover, the contribution of different electron–withdrawing groups on the benzothiazole ring influenced activity differently. (1) Br > Cl > NO2 > F (4d (43.37%, IC50 = 64.83 ± 4.20 μM) > 4b (39.27%, IC50 = 38.82 ± 3.76 μM) > 4n (12.51%, IC50 > 150 μM) > 4m (9.76%, IC50 > 150 μM)). (2) At the same time, substituting the halogen on the benzothiazole ring influenced the MAO inhibitory activity significantly. The ortho– or meta– position > para–position (Cl substituent 4b > 4c (21.59%, IC50 = 45.90 ± 2.66 μM), Br substituent 4d > 4f (24.15%, IC50 = 41.78 ± 2.94 μM)). Thus, substituents at the para–position reduced the inhibitory effect of compounds toward MAO. (3) Comparing the activities of mono– and di–substituted compounds with Cl or Br revealed that the di–Cl2 or di–Br2 substitution is not conducive to inhibiting MAO (Cl substituent 4b > 4o (10.19%, IC50 = 67.80 ± 1.26 μM), Br substituent 4d > 4p (19.80%, IC50 = 59.34 ± 3.22 μM)). According to the above–mentioned SAR results, we selected seven active target compounds to study the alternative inhibition of MAO–A and MAO–B further.

2.2.2. Selective Inhibition of MAO–A and MAO–B by the Derivatives

The inhibitory activities of derivatives (4a4p) against monoamine oxidase A (MAO–A) and MAO–B were evaluated with pagiline and clogiline as positive controls. The results are listed in Table 2. The six target compounds had negligible inhibitory activity against MAO–A. In contrast, the analogs all exhibited varying degrees of MAO–B antagonism. Among the six compounds, 4d had the highest inhibitory effect on MAO–A, but only an inhibition rate of 18.05%. Thus, this series of compounds may not have an antidepressant activity or exert antidepressant activity by other mechanisms. The specific results need to be further verified and confirmed by in vivo experiments. In particular, 4i (61.17%, IC50 = 16.49 ± 3.59 μM) displayed the strongest MAO–B inhibitory activity. Inhibition of MAO–B by 4g (IC50 = 12.12 ± 3.47 μM) was more significant, with the inhibition rate reaching 51.39%. However, the inhibition rate of 4g was not as strong as the positive control pargyline.
SAR data of benzothiazole–isoquinoline derivatives toward MAO–B are shown in Table 2. Based on the most active compound 4i, the introduction of the p–CH3 group on the benzothiazole ring is beneficial for inhibiting MAO–B, and the inhibitory activity of MAO–B is also very significant. Introducing the –CH3 at the para–position yielded stronger inhibition than placing this moiety at the ortho–position. The introduction of different electron–withdrawing groups had different effects on the MAO–B antagonism of target compounds: (1) Cl > Br, 4c (61.17%, IC50 = 9.13 ± 4.17 μM) > 4f (61.17%, IC50 = 47.61 ± 1.82 μM). (2) At the same time, substitution of the halogen on the benzothiazole ring also greatly affected the MAO–B inhibitory activity. With the para–position > meta– or ortho–position, (Cl substituent 4c (48.45%, IC50 = 9.13 ± 4.17 μM) > 4b (39.15%, IC50 = 40.84 ± 2.67 μM); Br substituent 4f (46.49%, IC50 = 47.61 ± 1.82 μM) > 4d (46.00%, IC50 = 3.26 ± 2.78 μM)). In addition, the benzothiazole–isoquinoline derivatives exhibited remarkable selectivity, demonstrating that they are MAO–B inhibitors with high potency and selectivity.

2.2.3. ChE Inhibition by the Benzothiazole–Isoquinoline Derivatives

The inhibitory activities of all benzothiazole–isoquinoline derivatives (4a4p) against BuChE were assessed in vitro by the modified Ellman method [57], and the positive control was tacolin. The results showed that a series of benzothiazole–isoquinoline derivatives showed no inhibitory effect on AChE. The majority of the benzothiazole–isoquinoline compounds demonstrated significant BuChE inhibitory activity because of the intricate design of the pharmacophore fusion strategy. Among them, compound 4d with o–Br as the substituent had the highest activity, and the inhibition rate was 77.76%; however, its inhibitory activity was far weaker than that of the positive control tacolin (99.41%, IC50 = 14.61 ± 5.81 μM).
The SAR results of benzothiazole–isoquinoline derivatives on BuChE are listed in Table 3. Different substitution positions displayed different anti–BuChE activities when the benzene ring was an electron–withdrawing substituent. The determined activity sequence was o–Br > m–Cl > m–Br > p–Cl > 2,6–Cl2 > 2,6–Br2 > o–Cl > p–Br > p–F > p–NO2. Compound 4d with o–Br as the substituent was observed to have the highest activity. The effect of the substitution position on activity differs when the benzene ring is an electron–donating substituent. The determined activity sequence was o–CH3 > m–OCH3 > o–OCH3 > p–CH3 > p–OCH3 > m–CH3. A comparison of the abilities of mono– and di–substituted compounds with Cl or Br was conducted: di–Cl2 or di–Br2 substitution was not conducive to inhibiting BuChE. (Cl substituent 4b (69.44%, IC50 = 17.59 ± 1.78 μM) > 4o (52.16%, IC50 = 21.76 ± 2.85 μM); Br substituent 4d (77.76%, IC50 = 14.61 ± 5.81 μM) > 4p (43.84%, IC50 = 27.12 ± 1.73 μM)).

2.3. In Vivo Biological Evaluation

The antidepressant activities of fluoxetine and benzothiazole–isoquinoline derivatives, as measured by the immobility time in the FST, are listed in Table 4. Most benzothiazole–isoquinoline derivatives induced a considerable reduction in the immobility time, suggesting that the series of derivatives may have antidepressant activity, but not through the monoamine oxidase mechanism. Specifically, 4d and 4g exhibited the strongest antidepressant effects, resulting in a considerable reduction in immobility duration in comparison to the control group (p < 0.001).
Compounds 4d and 4g reduced the immobility duration and displayed stronger %DID (percentage decrease in immobility duration) values than the other examine derivatives. The %DID values of 4d (65.06%) and 4g (61.32%) were slightly lower than those of fluoxetine (82.2%) at a concentration of 30 mg/kg in the FST. However, compared with the control group, all seven derivatives had significant antidepressant effects and their %DIDs were all greater than 50%.

2.4. Cellular Toxicity

Six benzothiazole–isoquinoline derivatives (4b, 4c, 4d, 4f, 4g, 4i) with strong activity were tested for cytotoxicity. The results of the MTT experiment are presented in Figure 2. Compared with the blank control group, the test results revealed that at 0–100 μM, the viability of the six benzothiazole–isoquinoline derivatives on cells all reached more than 90% and only showed weak toxicity at high concentrations.
Through the Acridine Orange (AO) fluorescence staining experiment, compared with the blank control group, the test results revealed that at 100 μM, the six benzothiazole–isoquinoline derivatives had no significant difference in cell viability, and the results are shown in Figure S1.

2.5. Molecular Docking

Molecular modeling was performed using the Vina (Simina) docking program to further investigate the interaction mode of compound 4g with BuChE and MAO–B (Figure 3). The interaction modes of compound 4g with BuChE and MAO–B revealed that derivative 4g binds to the active sites of BuChE and MAO–B, occupying the entire enzymatic CAS, mid–gorge site and PAS [58,59]. The docking results showed that 4g produced favorable binding energy values for both enzymes, with the greatest result observed for BuChE (−11.1 kcal·mol−1) and MAO–B (−9.2 kcal·mol−1). The blood–brain barrier (BBB) value for 4g was 0.84 suggesting the relatively good passage of this compound across the BBB. Upon docking with BuChE, the tetrahydroisoquinoline moiety binds to PAS, forming a π–π stacking interaction with Trp82 and Tyr332, whereas the amide moiety forms a hydrogen bond with Thr120. Upon docking with MAO–B, the amide moieties form hydrogen bonds with Gln206 and Ile199. These results indicate that the derivatives are dual inhibitors of BuChE and MAO–B, which is consistent with the results obtained from previous analyses.

3. Experimental Protocols

Chemical reagents were obtained and used directly from standard suppliers. Use Agilent liquid chromatograph to detect its purity. Column temperature: 35 °C, detection wavelength: 204 nm, mobile phase: methanol: water (V:V, 82:18), chromatographic column: DIAMONSILTM C18 column (250 mm × 4.6 mm, 5 μm).

3.1. The Synthetic Routes of Novel Benzothiazole–Isoquinoline Derivatives

3.1.1. General Procedure for the Synthesis of Substituted Benzo[d]thiazol–2–amine (2a2p)

In total, 50 mmol of substituted aniline (1a1p) and 250 mmol of KSCN were added to about 15 mL of acetic acid. Under ice bath conditions, glacial acetic acid mixed with 75 mmol of liquid bromine was slowly added dropwise, then at room temperature (RT) for 4 h. The reaction solution was poured into ice water to adjust pH. After the solid was precipitated, suction was filtered, the solid was dried, and recrystallized with ethanol to obtain the pure substituted benzo[d]thiazol–2–amine(2a2p).

3.1.2. General Procedure for the Synthesis of Substituted N–(benzo[d]thiazol–2–yl)–2–chloroacetamide (3a3p)

Dissolved 50 mmol of substituted benzo[d]thiazol–2–amine (2a2p) in tetrahydrofuran (THF), added 75 mmol of triethylamine, slowly added chloroacetyl chloride dropwise under ice bath conditions and stirred at RT for 1 h. To remove trimethylamine hydrochloride, the solvent was evaporated at decreased pressure and the residue was washed with water. The residue was dried and recrystallized to give the product substituted N–(benzo[d]thiazol–2–yl)–2–chloroacetamide (3a3p).

3.1.3. General Procedure for the Synthesis of Substituted (R)–N–(benzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4a4p)

Dissolved 50 mmol of substituted N–(benzo[d]thiazol–2–yl)–2–chloroacetamide (3a3p) in DMF, added 75 mmol of triethylamine and slowly added 75 mmol of (S)–1–phenyl–1,2,3,4–tetrahydroisoquinoline stirred in an oil bath under reflux for 1.5–2 h at 80 °C. The reaction solution was poured into ice water to adjust pH. After, the solid was precipitated, suction filtered and dried, and recrystallized to obtain pure (R)–N–(benzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4a4p).

3.2. The Spectral Information of Novel Benzothiazole–Isoquinoline Derivatives

3.2.1. (R)–N–(4–chlorobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4a)

HPLC/Purity: 91.6% (tR = 9.089), yield: 82.63%. mp: 245.6–247.0 °C.IR (KBr) cm−1: 3288, 1619, 1520, 1225.1H NMR(CDCl3, 300 MHz): δ 9.98 (1H, s, –NH), 7.33–7.12 (12H, m, –C6H5), 5.69 (1H, s, –CH), 4.86 (2H, t, –CH2–), 3.58–3.49 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 139.50, 138.98, 138.88, 138.67, 137.92, 137.74, 137.06, 136.38, 136.17, 135.86, 71.83, 68.46, 59.01, 39.01.ESI–MS calcd for C24H21ClN3OS+([M+H]+): 433.1097; found: 433.1078.

3.2.2. (R)–N–(5–chlorobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4b)

HPLC/Purity: 100.0% (tR = 10.829), yield: 81.48%. mp: 248.1–250.9 °C.IR (KBr) cm−1: 3287, 1617, 1515, 1228.1H NMR(CDCl3, 300 MHz): δ 9.18 (1H, s, –NH), 7.58–6.68 (12H, m, –C6H5), 5.28 (1H, s, –CH), 4.72 (2H, t, –CH2), 3.36–3.30 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 169.03, 142.60, 138.78, 137.39, 134.70, 133.62, 130.04, 129.55, 128.95, 128.82, 128.57, 128.10, 126.59, 126.22, 124.17, 119.38, 117.28, 77.30, 77.05, 76.79, 68.56, 58.95, 49.58, 29.40. ESI–MS calcd for C24H21ClN3OS+([M+H]+): 433.1097; found: 433.1078.

3.2.3. (R)–N–(6–chlorobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4c)

HPLC/Purity: 92.0% (tR = 11.526), yield: 80.17%. mp: 240.7–243.1 °C.IR (KBr) cm−1: 3280, 1615, 1513, 1224.1H NMR(CDCl3, 300 MHz): δ 12.11 (1H, s, –NH), 7.71–7.01 (12H, m, –C6H5), 4.98 (1H, s, –CH), 4.72 (2H, t, –CH2–), 2.98–2.84 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 170.83, 158.79, 147.88, 143.75, 138.33, 134.75, 133.70, 129.93, 129.02, 128.14, 127.89, 126.45, 126.11, 66.52, 57.04, 48.29, 29.24. ESI–MS calcd for C24H21ClN3OS+([M+H]+): 433.1097; found: 433.1078.

3.2.4. (R)–N–(4–bromobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4d)

HPLC/Purity: 100.0% (tR = 18.273), yield: 82.66%. mp: 254.3–255.9 °C.IR (KBr) cm−1: 3282, 1619, 1515, 1223.1H NMR(CDCl3, 300 MHz): δ 7.65 (1H, s, –NH), 7.64–6.61 (12H, m, –C6H5), 4.72 (1H, s, –CH), 4.42 (2H, t, –CH2–), 3.65–2.72 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 171.11, 146.46, 142.72, 136.91, 129.47, 129.02, 128.83, 128.77, 128.63, 128.51, 128.29, 126.32, 126.19, 126.02, 125. 95, 125.66, 116.22, 77.31, 77.26, 77.05, 76.80, 48.02, 43.48, 29.73, 17.57. ESI–MS calcd for C24H21BrN3OS+([M+H]+): 477.0573; found: 477.0578.

3.2.5. (R)–N–(5–bromobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4e)

HPLC/Purity: 100.0% (tR = 6.034), yield: 79.03%. mp: 255.3–256.9 °C.IR (KBr) cm−1: 3280, 1611, 1511, 1227.1H NMR(CDCl3, 300 MHz): δ 9.30 (1H, s, –NH), 7.97–6.70 (12H, m, –C6H5), 6.69 (1H, s, –CH), 4.73 (2H, t, –CH2–), 3.38–3.14 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 169.35, 142.47, 139.63, 137.16, 133.50, 131.30, 129.52, 128.95, 128.87, 128.58, 128.20, 126.69, 126.30, 123.81, 120.29, 119.44, 109.89, 77.29, 77.24, 77.03, 76.78, 68.63, 59.04, 49.67, 29.35. ESI–MS calcd for C24H21BrN3OS+([M+H]+): 477.0573; found: 477.0578.

3.2.6. (R)–N–(6–bromobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4f)

HPLC/Purity: 93.5% (tR = 19.415), yield: 77.40%. mp: 252.6–254.3 °C.IR (KBr) cm−1: 3289, 1611, 1517, 1220.1H NMR(CDCl3, 300 MHz): δ 10.35 (1H, s, –NH), 7.91–7.15 (12H, m, –C6H5), 6.67 (1H, s, –CH), 4.73 (2H, t, –CH2–), 3.48–2.86 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 169.79, 157.28, 147.51, 142.03, 137.25, 133.46, 129.72, 129.67, 128.93, 128.81, 128.53, 128.24, 126.66, 126.18, 123.93, 122.29, 116.94, 77.27, 77.02, 76.76, 69.03, 58.24, 50.37, 29.38. ESI–MS calcd for C24H21BrN3OS+([M+H]+): 477.0573; found: 477.0578.

3.2.7. (R)–N–(4–methylbenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4g)

HPLC/Purity: 93.5% (tR = 10.537), yield: 80.78%. mp: 247.6–249.7 °C.IR (KBr) cm−1: 3287, 1612, 1514, 1229.1H NMR(CDCl3, 300 MHz): δ 9.28 (1H, s, –NH), 7.37–7.02 (12H, m, –C6H5), 6.68 (1H, s, –CH), 4.71 (2H, t, –CH2–), 3.47–2.82 (4H, m, –C6H5), 1.56 (3H, s, –CH3) 13C NMR (CDCl3, 75 MHz):δ 169.04, 142.77, 137.80, 137.49, 133.39, 132.82, 130.04, 129.56, 128.93, 128.86, 128.52, 128.40, 128.19, 126.56, 126.27, 121.60, 117.69, 77.27, 77.02, 76.77, 68.69, 59.20, 50.13, 29.70, 17.80. ESI–MS calcd for C25H24N3OS+([M+H]+): 413.1673; found: 413.1678.

3.2.8. (R)–N–(5–methylbenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4h)

HPLC/Purity: 98.5% (tR = 9.147), yield: 78.27%. mp: 241.3–243.6 °C.IR (KBr) cm−1: 3290, 1617, 1514, 1226.1H NMR(CDCl3, 300 MHz): δ 9.26 (1H, s, –NH), 7.54–7.04 (12H, m, –C6H5), 6.79 (1H, s, –CH), 4.73 (2H, t, –CH2–), 3.37–3.15 (4H, m, –C6H5), 1.56 (3H, s, –CH3),13C NMR (CDCl3, 75 MHz):δ 169.29, 142.52, 141.63, 139.90, 137.30, 134.23, 133.57, 129.53, 129.96, 128.82, 128.57, 128,12, 126.62, 126.25, 121.64, 118.17, 117.26, 110.72, 77.27, 77.02, 76.76, 68.56, 59.01, 49.60, 29.37, 20.83. ESI–MS calcd for C25H24N3OS+([M+H]+): 413.1673; found: 413.1678.

3.2.9. (R)–N–(6–methylbenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4i)

HPLC/Purity: 95.1% (tR = 15.277), yield: 76.96%. mp: 244.8–246.9 °C.IR (KBr) cm−1: 3288, 1612, 1519, 1220.1H NMR(CDCl3, 300 MHz): δ 10.35 (1H, s, –NH), 7.70–7.04 (12H, m, –C6H5), 6.69 (1H, s, –CH), 4.79 (2H, t, –CH2–), 3.28–2.84 (4H, m, –C6H5), 1.66 (3H, s, –CH3), 13C NMR (CDCl3, 75 MHz):δ 169.59, 159.23, 146.38, 142.10, 137.33, 134.00, 133.53, 132.37, 129.73, 129.54, 129.50, 128.88, 128.78, 128.74, 128.52, 128.17, 127.72, 126.59, 126.10, 121.19, 120.59, 119.40, 77.45, 77.02, 76.60, 68.99, 58.26, 50.31, 29.36, 21.47. ESI–MS calcd for C25H24N3OS+([M+H]+): 413.1673; found: 413.1678.

3.2.10. (R)–N–(4–methoxybenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4j)

HPLC/Purity: 93.1% (tR = 10.766), yield: 75.88%. mp: 253.7–254.9 °C.IR (KBr) cm−1: 3281, 1610, 1516, 1222.1H NMR(CDCl3, 300 MHz): δ 9.91 (1H, s, –NH), 7.76–6.69 (12H, m, –C6H5), 4.72 (1H, s, –CH), 3.95 (2H, t, –CH2–), 3.37–2.82 (4H, m, –C6H5), 1.26 (3H, s, –CH3), 13C NMR (CDCl3, 75 MHz):δ 169.31, 149.08, 142.86, 137.70, 133.77, 129.48, 128.92, 128.70, 128.51, 127.97, 126.41, 126.12, 124.64, 120.27, 117.10, 112.69, 111.09, 77.29, 77.04, 76.78, 68.49, 59.03, 56.25, 49.75, 29.52. ESI–MS calcd for C25H24N3O2S+([M+H]+): 429.1573; found: 429.1578.

3.2.11. (R)–N–(5–methoxybenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4k)

HPLC/Purity: 91.4% (tR = 8.686), yield: 73.37%. mp: 248.9–250.1 °C.IR (KBr) cm−1: 3288, 1615, 1512, 1229.1H NMR(CDCl3, 300 MHz): δ 9.91 (1H, s, –NH), 7.31–6.71 (12H, m, –C6H5), 5.29 (1H, s, –CH), 4.72 (2H, t, –CH2–), 3.95–3.08 (4H, m, –C6H5), 2.80 (3H, s, –CH3), 13C NMR (CDCl3, 75 MHz):δ 160.24, 138.88, 133.71, 129.71, 129.55, 128.96, 128.78, 128.57, 128.03, 126.54, 126.17, 111.50, 110.00, 105.07, 77.28, 77.03, 76.78, 68.49, 58.95, 55.35, 49.48, 29.37. ESI–MS calcd for C25H24N3O2S+([M+H]+): 429.1573; found: 429.1578.

3.2.12. (R)–N–(6–methoxybenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4l)

HPLC/Purity: 93.3% (tR = 13.066), yield: 75.24%. mp: 247.9–249.4 °C.IR (KBr) cm−1: 3286, 1617, 1514, 1229.1H NMR(CDCl3, 300 MHz): δ 9.15 (1H, s, –NH), 7.33–7.16 (12H, m, –C6H5), 6.69 (1H, s, –CH), 4.73 (2H, t, –CH2–), 3.80–3.09 (4H, m, –C6H5), 1.58 (3H, s, –CH3), 13C NMR (CDCl3, 75 MHz):δ 169.44, 156.87, 142.70, 142.11, 137.33, 133.53, 133.50, 129.73, 128.89, 128.80, 128.53, 128.18, 126.60, 126.12, 121.63, 115.23, 104.31, 77.30, 77.25, 77.04, 76.79, 68.99, 58.22, 55.84, 50.29, 29.37, 14.13. ESI–MS calcd for C25H24N3O2S+([M+H]+): 429.1573; found: 429.1578.

3.2.13. (R)–N–(6–fluorobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4m)

HPLC/Purity: 92.0% (tR = 11.526), yield: 86.40%. mp: 255.6–258.1 °C.IR (KBr) cm−1: 3280, 1616, 1513, 1227.1H NMR(CDCl3, 300 MHz): δ 11.98 (1H, s, –NH), 7.83–6.95 (12H, m, –C6H5), 4.92 (1H, s, –CH), 3.37 (2H, t, –CH2–), 2.92–2.44 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 157.64, 129.40, 128.51, 128.18, 127.37, 125.76, 121.63, 114.22, 108.17, 66.00, 56.49, 47.77, 28.72.ESI–MS calcd for C24H21FN3OS+([M+H]+): 417.1373; found: 417.1378.

3.2.14. (R)–N–(6–nitrobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4n)

HPLC/Purity:90.8% (tR = 6.333), yield: 82.59%. mp: 240.6–243.2 °C.IR (KBr) cm−1: 3283, 1610, 1517, 1223.1H NMR(CDCl3, 300 MHz): δ 12.43 (1H, s, –NH), 7.84–6.98 (12H, m, –C6H5), 6.62 (1H, s, –CH), 4.96 (2H, t, –CH2–), 3.15–2.84 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 170.87, 162.99, 153.39, 143.09 (d, J = 29.3 Hz), 137.80, 134.23, 129.41, 128.50, 128.37, 128.27, 127.38, 125.94, 125.60, 121.76, 120.57, 119.03, 66.00, 56.60, 47.79, 28.73.ESI–MS calcd for C24H21N4O3S+([M+H]+): 444.1373; found: 444.1378.

3.2.15. (R)–N–(4,6–dichlorobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4o)

HPLC/Purity: 94.8% (tR = 22.278), yield: 71.83%. mp: 253.5–256.1 °C.IR (KBr) cm−1: 3287, 1614, 1516, 1223.1H NMR(CDCl3, 300 MHz): δ 9.87 (1H, s, –NH), 8.42–6.67 (12H, m, –C6H5), 5.29 (1H, s, –CH), 4.70 (2H, t, –CH2–), 3.45–2.84 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 169.24, 133.63, 133.46, 129.61, 128.85, 128.82, 128.72, 128.51, 128.06, 127.88, 126.41, 126.11, 121.33, 77.28, 77.23, 77.02, 76.77, 68.78, 59.17, 50.33, 29.70. ESI–MS calcd for C24H21Cl2N3OS+([M+H]+): 467.0673; found: 467.0678.

3.2.16. (R)–N–(4,6–dibromobenzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamide (4p)

HPLC/Purity: 93.0% (tR = 10.816), yield: 73.12%. mp: 258.6–260.1 °C.IR (KBr) cm−1: 3280, 1612, 1510, 1221.1H NMR(CDCl3, 300 MHz): δ 9.86 (1H, s, –NH), 7.91–7.03 (12H, m, –C6H5), 6.60 (1H, s, –CH), 4.83 (2H, t, –CH2–), 3.25–2.78 (4H, m, –C6H5), 13C NMR (CDCl3, 75 MHz):δ 178.73, 152.94, 147.56, 144.13, 143.76, 141.22, 139.45, 138.36, 137.56, 135.96, 135.68, 132.14, 59.14, 38.94. ESI–MS calcd for C24H21Br2N3OS+([M+H]+): 554.9673; found: 554.9678.

3.3. Determination of the Inhibitory Activity of 4a4p on MAO by Holts Method

3.3.1. Preparation of MAO

Male SD rats were sacrificed by decapitation, operated on ice, their livers were removed, and the liver surface was washed with precooled phosphate buffer. The liver was cut into pieces, and after washing (using PBS), 20 mL of 0.3 M sucrose buffer was added for homogenization. After fully homogenized, it was balanced by a tray balance. Low–temperature differential centrifugation was performed, and the obtained SD rat liver mitochondrial concentrate was the experimental monoamine oxidase stock solution, and was stored in a −80 °C refrigerator.

3.3.2. MAO Inhibitory Activity and IC50 Detection

Added 25 µL of enzyme dilution solution and 25 µL of test derivatives (control group plus rasagiline, blank group plus 10% DMSO) to 96–well plate, incubated at 37 °C for 1 h, then 120 µL of substrate was added, 80 µL of chromogenic solution was added, incubated at 37 °C for 30 min, and its absorbance measured at 490 nm. In addition, in order to exclude the influence of protein, a protein control group was established, only the enzyme dilution was added, and the rest were replaced with PBS, and the absorbance value was detected at a wavelength of 490 nm. The inhibition rate of each concentration was substituted into the calculation software GraphPad Prism 8 to calculate the IC50 value.

3.3.3. MAO–A Inhibitory Activity and IC50 Detection

Added 25 µL of enzyme solution and 25 µL of 10 μM pargyline solution to the 96–well plate in sequence, and incubated at 37 °C for 30 min, other operations were the same as 4.3.2 Monoamine Oxidase Inhibitory Activity and IC50 Detection. The inhibition rate of each concentration was substituted into the calculation software GraphPad Prism 8 to calculate the IC50 value.

3.3.4. MAO–B Inhibitory Activity and IC50 Detection

Added 25 µL of enzyme solution and 25 µL of 10 μM chlorgiline solution to the 96–well plate in sequence, and incubated at 37 °C for 30 min, other operations were the same as 4.3.2 Monoamine Oxidase Inhibitory Activity and IC50 Detection. The inhibition rate of each concentration was substituted into the calculation software GraphPad Prism 8 to calculate the IC50 value.

3.4. Determination of the Inhibitory Activity of Target Compounds 4a4p on ChE

Compounds 4a4p were dissolved in DMSO and then diluted in 10% DMSO to obtain sample solutions of different concentrations. Each concentration was tested three times in parallel, and 96 empty plates were taken, and 20 µL of the test sample or positive drug (10% DMSO was added to the blank control), 40 µL of AChE or BuChE (0.2 U/mL) and 100 µL of DTNB (0.001 mol/L) were added in turn, mixed well and incubated in a constant temperature water bath at 37 °C for 15 min. Then, 20 µL of the substrate ATCI or BTCI (0.001 mol/L) was added, and after mixing evenly, the mixture was incubated in a constant temperature water bath at 37 °C for 3 min, and the absorbance value was measured at 412 nm.

3.5. Forced Swimming Test

Pharmacological experiments of antidepressant activity used FST in mice, fluoxetine hydrochloride was used as a positive control, and the vehicle solvent PFG–400 was used as a blank control. The experiment used ICR male mice (20 ± 2 g). Six benzothiazole–quinoline derivatives were dissolved in polyethylene glycol–400, and 30 min after administration (i.p., 30 mg/kg), the mice were each placed in a glass cylinder, recording the time of immobility. Duration of immobility outcomes is expressed as mean ± SEM. The t-test was used to make comparisons between groups.

3.6. Cell Culture

L929 (Shanghai Cell Bank, Chinese Academy of Sciences) cells were cultured in 10% fetal bovine serum: penicillin–streptomycin solution: MEM mixed medium with serum–free medium (10:1:90), and subcultured in a 5% carbon dioxide cell incubator at 37 °C, and the medium changed every other day.

3.7. Assessment of Cytotoxicity

In this experiment, L929 cells in the logarithmic growth phase were selected, and the cell density was adjusted to 2 × 104 cells per well. The cells were seeded in 96–well plates, and the 96–well plates seeded with cells were cultured in a 37 °C 5% carbon dioxide cell incubator. After 24 h, different concentrations of the test compound medium (6.5, 12.5, 25, 50, 100 μM) were added, and the blank group was added with drug–free medium, shaken gently, and placed at 37 °C 5%. The cells were cultured in a carbon dioxide cell incubator for 24 h, and each concentration was replicated 6 times. After 24 h, 20 μL of 5 mg/kg MTT solution (prepared in PBS with pH = 7.6) was added in the dark, and cultured in the cell incubator for another 4 h. After 4 h, the 96–well plate was taken out and the original medium was discarded. Added 160 μL DMSO, put it on a shaker and mixed well for about 10 min, used a microplate reader to measure its absorbance at 490 nm, recorded the results and analyzed.

3.8. Analyze Cell Viability by AO Staining

L929 cells in the logarithmic growth phase were selected, and the cell density was adjusted to 8 × 105 per well with a cell counter to inoculate in a 6–well plate, and the 6–well plate inoculated with cells was cultured in a 37 °C 5% carbon dioxide cell incubator. After 24 h, 1 mL of medium containing 100 μM of the compound to be tested was added (medium without drug was added to the blank group), shaken gently, and cultured in a 37 °C 5% carbon dioxide cell incubator. After 8 h, the medium was discarded, PBS was added to wash once, 1 mL of PBS and 80 μL of AO solution were slowly added in the dark, mixed for 5 min to make the staining uniform and sufficient, after 5 min, the stain was aspirated and washed twice with PBS, and observed under a fluorescence microscope, the results saved and analyzed.

3.9. Molecular Docking

Molecular simulations and docking experiments were performed using the Vina (Simina) docking program. The protein crystal structures of human MAO–B and BuChE were obtained from the Protein Crystal Database (PDBID: 2Z5X and 4BDS), resolution is 5Å (10−10 m). It mainly simulates the docking process between inhibitors and MAO–B and BuChE. Compound 4g was selected as the inhibitor target for molecular modeling, and the initial structures of MAO–B and BuChE protein crystals were processed with default parameters, the pocket is the FAD ligand center, molecular simulations were performed between the optimized protein crystal structure and compound 4g. The inhibitory effect was evaluated at the molecular level, which provided ideas for the design of better MAO and BuChE inhibitors in the future.

4. Conclusions

In this report, we designed and synthesized a series of (R)–N–(benzo[d]thiazol–2–yl)–2–(1–phenyl–3,4–dihydroisoquinolin–2(1H)–yl)acetamides as new multi–target inhibitors that potentially display multi–functional anti–neurodegenerative and antidepressant activities. Substituents on the benzene ring of benzothiazole were changed to investigate their effects on the selective inhibitory activities against MAO–B and BuChE, and SAR analysis was carried out. The inhibitory effect of benzothiazole–isoquinoline derivatives on MAO–A was found to be substantially less than against MAO–B. Compound 4d displayed a mild inhibitory effect toward MAO–A, whereas six derivatives reduced MAO–B activity significantly. Compounds 4g and 4i gave the strongest inhibitory activity toward MAO–B. All derivatives exhibited selective inhibition of BuChE, with 4b4e and 4g displaying the strongest inhibition. The FST showed that this series of benzothiazole–isoquinoline derivatives (4d and 4g) reduced the immobility time significantly. This observation indicates that this series of compounds does have antidepressant potential, but not through inhibition of monoamine oxidase and choline, and the specific mechanism of action needs to be explored further. Subsequently, their cytotoxicity was assessed by the MTT assay and AO staining, and the results showed that the compounds were not toxic to L929 cells at effective concentrations. Molecular docking studies were carried out for 4g to define the possible binding mode of this compound. The primary interaction site between the compound and the two enzymes was revealed, and was consistent with the observed in vitro MAO–B and BuChE inhibitory activities of these compounds, thereby broadening our understanding of the requirements for the compounds to achieve high affinity. The above results suggest that compound 4g with multi–targeting is a promising drug candidate for treating neurodegenerative diseases complicated by depression. Nonetheless, further research on the disease pathways and pathophysiology of neurodegenerative diseases complicated by depression is required to clarify the pathogenesis of the disease and in the development of preventive and therapeutic measures.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules27249062/s1, Figure S1: The effect of benzothiazole-isoquinoline derivatives (4b4d, 4f, 4g, 4i) on the proliferation of L929 cells; Figure S2–S17: HPLC Data of Derivatives (4a4p); Figure S18–S49: NMR Data of Derivatives (4a4p).

Author Contributions

Conceptualization, L.G. and W.L.; data curation, W.L.; formal analysis, W.L.; funding acquisition, S.W. and L.G.; investigation, W.L.; methodology, W.L. and Z.H.; project administration, Z.H.; resources, D.Z.; software, W.L., L.Z. and Y.Z.; supervision, D.Z., L.J., L.G. and S.W.; validation, W.L., Y.H. and S.W.; visualization, Y.H.; writing—original draft, W.L. and D.Z.; writing—review and editing, L.G. and S.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Committee on the Ethics of Animal Experiments of the Zhejiang Ocean University (Permit Number:20135107G).

Data Availability Statement

The data presented in this study and associated additional data are available upon request.

Acknowledgments

This was provided by Zhejiang Ocean University Undergraduate Innovation Project (Academic Papers, 2022) and the Province of Jilin Science and Technology Development Plan (20210502028ZP), and the National Natural Science Foundation of China (No. 21964017).

Conflicts of Interest

The authors declare that they have no known competing financial interest or personal relationship that could have appeared to influence the work reported in this paper.

References

  1. Gitler, A.D.; Dhillon, P.; Shorter, J. Neurodegenerative Disease: Models, Mechanisms, and a New Hope. DMM Dis. Model. Mech. 2017, 10, 499–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. De Tommaso, M.; Arendt–Nielsen, L.; Defrin, R.; Kunz, M.; Pickering, G.; Valeriani, M. Pain in Neurodegenerative Disease: Current Knowledge and Future Perspectives. Behav. Neurol. 2016, 2016, 7576292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a Risk Factor for Neurodegenerative Disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
  4. Duman, R.S.; Sanacora, G.; Krystal, J.H. Altered Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron 2020, 102, 75–90. [Google Scholar] [CrossRef]
  5. Iqbal, K.; Grundke–Iqbal, I. Alzheimer Disease Is Multifactorial and Heterogeneous. Neurobiol. Aging 2000, 21, 901–902. [Google Scholar] [CrossRef]
  6. Ballard, C.; Gauthier, S.; Corbett, A.; Brayne, C.; Aarsland, D.; Jones, E. Alzheimer’s Disease. Lancet 2011, 377, 1019–1031. [Google Scholar] [CrossRef]
  7. Anand, R.; Gill, K.D.; Mahdi, A.A. Therapeutics of Alzheimer’s Disease: Past, Present and Future. Neuropharmacology 2014, 76, 27–50. [Google Scholar] [CrossRef]
  8. Zhao, Q.F.; Tan, L.; Wang, H.F.; Jiang, T.; Tan, M.S.; Tan, L.; Xu, W.; Li, J.Q.; Wang, J.; Lai, T.J.; et al. The Prevalence of Neuropsychiatric Symptoms in Alzheimer’s Disease: Systematic Review and Meta–Analysis. J. Affect. Disord. 2016, 190, 264–271. [Google Scholar] [CrossRef]
  9. Mroueh, M.; Faour, W.H.; Shebaby, W.N.; Daher, C.F.; Ibrahim, T.M.; Ragab, H.M. Synthesis, Biological Evaluation and Modeling of Hybrids from Tetrahydro–1H–Pyrazolo[3,4–b]Quinolines as Dual Cholinestrase and COX–2 Inhibitors. Bioorg. Chem. 2020, 100, 103895. [Google Scholar] [CrossRef]
  10. Guo, Z.; Zhang, J.; Liu, X.; Hou, H.; Cao, Y.; Wei, F.; Li, J.; Chen, X.; Shen, Y.; Chen, W. Neurometabolic Characteristics in the Anterior Cingulate Gyrus of Alzheimer’s Disease Patients with Depression: A 1H Magnetic Resonance Spectroscopy Study. BMC Psychiatry 2015, 15, 306. [Google Scholar] [CrossRef]
  11. Zahodne, L.B.; Gongvatana, A.; Cohen, R.A.; Ott, B.R.; Tremont, G. Are Apathy and Depression Independently Associated with Longitudinal Trajectories of Cortical Atrophy in Mild Cognitive Impairment? Am. J. Geriatr. Psychiatry 2013, 21, 1098–1106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Verma, A.; Kumar Waiker, D.; Bhardwaj, B.; Saraf, P.; Shrivastava, S.K. The Molecular Mechanism, Targets, and Novel Molecules in the Treatment of Alzheimer’s Disease. Bioorg. Chem. 2022, 119, 105562. [Google Scholar] [CrossRef] [PubMed]
  13. Santos, L.E.; Beckman, D.; Ferreira, S.T. Microglial Dysfunction Connects Depression and Alzheimer’s Disease. Brain. Behav. Immun. 2016, 55, 151–165. [Google Scholar] [CrossRef] [PubMed]
  14. Elsworthy, R.J.; Aldred, S. Depression in Alzheimer’s Disease: An Alternative Role for Selective Serotonin Reuptake Inhibitors? J. Alzheimer Dis. 2019, 69, 651–661. [Google Scholar] [CrossRef] [PubMed]
  15. Galts, C.P.C.; Bettio, L.E.B.; Jewett, D.C.; Yang, C.C.; Brocardo, P.S.; Rodrigues, A.L.S.; Thacker, J.S.; Gil–Mohapel, J. Depression in Neurodegenerative Diseases: Common Mechanisms and Current Treatment Options. Neurosci. Biobehav. Rev. 2019, 102, 56–84. [Google Scholar] [CrossRef]
  16. Kim, H.J.; Kim, W.; Kong, S.Y. Antidepressants for Neuro–Regeneration: From Depression to Alzheimer’s Disease. Arch. Pharm. Res. 2013, 36, 1279–1290. [Google Scholar] [CrossRef]
  17. Mitić, M.; Lazarević–Pašti, T. Does the Application of Acetylcholinesterase Inhibitors in the Treatment of Alzheimer’s Disease Lead to Depression? Expert Opin. Drug Metab. Toxicol. 2021, 17, 841–856. [Google Scholar] [CrossRef]
  18. Bergantin, L.B. The Interactions Between Alzheimer’s Disease and Major Depression: Role of Ca2+ Channel Blockers and Ca2+/CAMP Signalling. Curr. Drug Res. Rev. 2020, 12, 97–102. [Google Scholar] [CrossRef]
  19. Cummings, J.L.; Tong, G.; Ballard, C. Treatment Combinations for Alzheimer’s Disease: Current and Future Pharmacotherapy Options. J. Alzheimer Dis. 2019, 67, 779–794. [Google Scholar] [CrossRef] [Green Version]
  20. O’Leary, O.F.; Dinan, T.G.; Cryan, J.F. Faster, Better, Stronger: Towards New Antidepressant Therapeutic Strategies. Eur. J. Pharmacol. 2015, 753, 32–50. [Google Scholar] [CrossRef]
  21. de Picker, L.; van Den Eede, F.; Dumont, G.; Moorkens, G.; Sabbe, B.G.C. Antidepressants and the Risk of Hyponatremia: A Class–by–Class Review of Literature. Psychosomatics 2014, 55, 536–547. [Google Scholar] [CrossRef] [PubMed]
  22. Sultana, J.; Spina, E.; Trifirò, G. Antidepressant Use in the Elderly: The Role of Pharmacodynamics and Pharmacokinetics in Drug Safety. Expert Opin. Drug Metab. Toxicol. 2015, 11, 883–892. [Google Scholar] [CrossRef] [PubMed]
  23. Porsteinsson, A.P.; Drye, L.T.; Pollock, B.G.; Devanand, D.P.; Frangakis, C.; Ismail, Z.; Marano, C.; Meinert, C.L.; Mintzer, J.E.; Munro, C.A.; et al. Effect of Citalopram on Agitation in Alzheimer Disease: The CitAD Randomized Clinical Trial. JAMA–J. Am. Med. Assoc. 2014, 311, 682–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Gahr, M.; Zeiss, R.; Lang, D.; Connemann, B.J.; Hiemke, C.; Muche, R.; Freudenmann, R.W.; Schönfeldt–Lecuona, C. Association between Haemorrhages and Treatment with Selective and Non–Selective Serotonergic Antidepressants: Possible Implications of Quantitative Signal Detection. Psychiatry Res. 2015, 229, 257–263. [Google Scholar] [CrossRef]
  25. Jasiak, N.M.; Bostwick, J. RRisk of QT/QTc Prolongation Among Newer Non–SSRI Antidepressants. Ann. Pharmacother. 2014, 48, 1620–1628. [Google Scholar] [CrossRef]
  26. Lozupone, M.; La Montagna, M.; D’Urso, F.; Piccininni, C.; Sardone, R.; Dibello, V.; Giannelli, G.; Solfrizzi, V.; Greco, A.; Daniele, A.; et al. Pharmacotherapy for the Treatment of Depression in Patients with Alzheimer’s Disease: A Treatment–Resistant Depressive Disorder. Expert Opin. Pharmacother. 2018, 19, 823–842. [Google Scholar] [CrossRef]
  27. Siarkos, K.T.; Katirtzoglou, E.A.; Politis, A.M. A Review of Pharmacological Treatments for Depression in Alzheimer’s Disease. J. Alzheimer Dis. 2015, 48, 15–34. [Google Scholar] [CrossRef]
  28. Orgeta, V.; Tabet, N.; Nilforooshan, R.; Howard, R. Efficacy of Antidepressants for Depression in Alzheimer’s Disease: Systematic Review and Meta–Analysis. J. Alzheimer Dis. 2017, 58, 725–733. [Google Scholar] [CrossRef] [Green Version]
  29. Li, X.; Li, J.; Huang, Y.; Gong, Q.; Fu, Y.; Xu, Y.; Huang, J.; You, H.; Zhang, D.; Zhang, D.; et al. The Novel Therapeutic Strategy of Vilazodone–Donepezil Chimeras as Potent Triple–Target Ligands for the Potential Treatment of Alzheimer’s Disease with Comorbid Depression. Eur. J. Med. Chem. 2022, 229, 114045. [Google Scholar] [CrossRef]
  30. Wang, T.; Liu, X.H.; Guan, J.; Ge, S.; Wu, M.B.; Lin, J.P.; Yang, L.R. Advancement of Multi–Target Drug Discoveries and Promising Applications in the Field of Alzheimer’s Disease. Eur. J. Med. Chem. 2019, 169, 200–223. [Google Scholar] [CrossRef]
  31. Weller, J.; Budson, A. Current Understanding of Alzheimer’s Disease Diagnosis and Treatment. F1000Research 2018, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Deng, X.; Zhao, S.; Liu, X.; Han, L.; Wang, R.; Hao, H.; Jiao, Y.; Han, S.; Bai, C. Polygala Tenuifolia: A Source for Anti–Alzheimer’s Disease Drugs. Pharm. Biol. 2020, 58, 410–416. [Google Scholar] [CrossRef] [PubMed]
  33. Reis, J.; Cagide, F.; Valencia, M.E.; Teixeira, J.; Bagetta, D.; Pérez, C.; Uriarte, E.; Oliveira, P.J.; Ortuso, F.; Alcaro, S.; et al. Multi–Target–Directed Ligands for Alzheimer’s Disease: Discovery of Chromone–Based Monoamine Oxidase/Cholinesterase Inhibitors. Eur. J. Med. Chem. 2018, 158, 781–800. [Google Scholar] [CrossRef] [PubMed]
  34. Rodríguez–Soacha, D.A.; Scheiner, M.; Decker, M. Multi–Target–Directed–Ligands Acting as Enzyme Inhibitors and Receptor Ligands. Eur. J. Med. Chem. 2019, 180, 690–706. [Google Scholar] [CrossRef]
  35. Rossi, M.; Freschi, M.; De Camargo Nascente, L.; Salerno, A.; De Melo Viana Teixeira, S.; Nachon, F.; Chantegreil, F.; Soukup, O.; Prchal, L.; Malaguti, M.; et al. Sustainable Drug Discovery of Multi–Target–Directed Ligands for Alzheimer’s Disease. J. Med. Chem. 2021, 64, 4972–4990. [Google Scholar] [CrossRef]
  36. Guan, L.P.; Xia, Y.N.; Jin, Q.H.; Liu, B.Y.; Wang, S.H. Synthesis, Potential Anti–Inflammatory and Analgesic Activities Study of (S)–N–Substituted–1–Phenyl–3,4–Dihydroisoquinoline–2(1H)–Carboxamides. Bioorg. Med. Chem. Lett. 2017, 27, 3378–3381. [Google Scholar] [CrossRef]
  37. Zhen, X.; Peng, Z.; Zhao, S.; Han, Y.; Jin, Q.; Guan, L. Synthesis, Potential Anticonvulsant and Antidepressant Effects of 2–(5–Methyl–2,3–Dioxoindolin–1–Yl)Acetamide Derivatives. Acta Pharm. Sin. B 2015, 5, 343–349. [Google Scholar] [CrossRef] [Green Version]
  38. Jin, Q.; Fu, Z.; Guan, L.; Jiang, H. Syntheses of Benzo[d]Thiazol–2(3H)–One Derivatives and Their Antidepressant and Anticonvulsant Effects. Mar. Drugs 2019, 17, 430. [Google Scholar] [CrossRef] [Green Version]
  39. Guan, L.-P.; Liu, B.-Y.; Quan, Y.-C.; Yang, L.-Y.; Zhen, X.-H.; Wang, S.-H. Synthesis and Evaluation of Phenyliminoindolin–Containing Phenylacetamide Derivatives with the Antidepressant and Anticonvulsant Effects. Med. Chem. 2016, 12, 786–794. [Google Scholar] [CrossRef]
  40. Haider, K.; Rehman, S.; Pathak, A.; Najmi, A.K.; Yar, M.S. Advances in 2–Substituted Benzothiazole Scaffold–Based Chemotherapeutic Agents. Arch. Pharm. 2021, 354, 2100246. [Google Scholar] [CrossRef]
  41. Rouf, A.; Tanyeli, C. Bioactive Thiazole and Benzothiazole Derivatives. Eur. J. Med. Chem. 2015, 97, 911–927. [Google Scholar] [CrossRef] [PubMed]
  42. Erdogan, M.; Kilic, B.; Sagkan, R.I.; Aksakal, F.; Ercetin, T.; Gulcan, H.O.; Dogruer, D.S. Design, Synthesis and Biological Evaluation of New Benzoxazolone/Benzothiazolone Derivatives as Multi–Target Agents against Alzheimer’s Disease. Eur. J. Med. Chem. 2021, 212, 113124. [Google Scholar] [CrossRef] [PubMed]
  43. Wichur, T.; Więckowska, A.; Więckowski, K.; Godyń, J.; Jończyk, J.; Valdivieso, Á.D.R.; Panek, D.; Pasieka, A.; Sabaté, R.; Knez, D.; et al. 1–Benzylpyrrolidine–3–Amine–Based BuChE Inhibitors with Anti–Aggregating, Antioxidant and Metal–Chelating Properties as Multifunctional Agents against Alzheimer’s Disease. Eur. J. Med. Chem. 2020, 187, 111916. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, Y.; Cho, H.J.; Bandara, N.; Sun, L.; Tran, D.; Rogers, B.E.; Mirica, L.M. Metal–Chelating Benzothiazole Multifunctional Compounds for the Modulation and 64Cu PET Imaging of Aβ Aggregation. Chem. Sci. 2020, 11, 7789–7799. [Google Scholar] [CrossRef]
  45. Özkay, Ü.D.; Kaya, C.; Çevik, U.A.; Can, Ö.D. Synthesis and Antidepressant Activity Profile of Some Novel Benzothiazole Derivatives. Molecules 2017, 22, 1490. [Google Scholar] [CrossRef]
  46. Geerts, H.; Nuydens, R.; De Jong, M.; Cornelissen, F.; Nuyens, R.; Wouters, L. Sabeluzole Stabilizes the Neuronal Cytoskeleton. Neurobiol. Aging 1996, 17, 573–581. [Google Scholar] [CrossRef]
  47. Abe, K.; Saitoh, T.; Horiguchi, Y.; Utsunomiya, I.; Taguchi, K. Synthesis and Neurotoxicity of Tetrahydroisoquinoline Derivatives for Studying Parkinson’s Disease. Biol. Pharm. Bull. 2005, 28, 1355–1362. [Google Scholar] [CrossRef] [Green Version]
  48. Sampaio, T.B.; Da Rocha, J.T.; Prigol, M.; Saraiva, R.A.; Nogara, P.F.; Stein, A.L.A.; da Rocha, J.B.T.; Zeni, G.; Nogueira, C.W. 4–Organoseleno–Isoquinolines Selectively and Reversibly Inhibit the Cerebral Monoamine Oxidase B Activity. J. Mol. Neurosci. 2016, 59, 135–145. [Google Scholar] [CrossRef]
  49. Shang, X.F.; Yang, C.J.; Morris–Natschke, S.L.; Li, J.C.; Yin, X.D.; Liu, Y.Q.; Guo, X.; Peng, J.W.; Goto, M.; Zhang, J.Y.; et al. Biologically Active Isoquinoline Alkaloids Covering 2014–2018. Med. Res. Rev. 2020, 40, 2212–2289. [Google Scholar] [CrossRef]
  50. Liu, P.; Chen, X.; Zhou, H.; Wang, L.; Zhang, Z.; Ren, X.; Zhu, F.; Guo, Y.; Huang, X.; Liu, J.; et al. The Isoquinoline Alkaloid Dauricine Targets Multiple Molecular Pathways to Ameliorate Alzheimer–like Pathological Changes in Vitro. Oxid. Med. Cell. Longev. 2018, 2018, 2025914. [Google Scholar] [CrossRef]
  51. Możdżeń, E.; Wąsik, A.; Romańska, I.; Michaluk, J.; Antkiewicz–Michaluk, L. Antidepressant–like Effect of 1,2,3,4–Tetrahydroisoquinoline and Its Methyl Derivative in Animal Models of Depression. Pharmacol. Rep. 2017, 69, 566–574. [Google Scholar] [CrossRef] [PubMed]
  52. Abdel–Aziz, M.; Abuo–Rahma, G.E.D.A.; Hassan, A.A. Synthesis of Novel Pyrazole Derivatives and Evaluation of Their Antidepressant and Anticonvulsant Activities. Eur. J. Med. Chem. 2009, 44, 3480–3487. [Google Scholar] [CrossRef] [PubMed]
  53. Suryawanshi, M.R.; Kanhed, A.M.; Kulkarni, V.M.; Bhosale, S.H.; Yadav, M.R. Design, Synthesis, and Computational Studies of Phenylacetamides as Antidepressant Agents. Mol. Divers. 2022, 26, 3157–3172. [Google Scholar] [CrossRef] [PubMed]
  54. Kumari, S.; Carmona, A.V.; Tiwari, A.K.; Trippier, P.C. Amide Bond Bioisosteres: Strategies, Synthesis, and Successes. J. Med. Chem. 2020, 63, 12290–12358. [Google Scholar] [CrossRef] [PubMed]
  55. Begum, S.; Nizami, S.S.; Mahmood, U.; Masood, S.; Iftikhar, S.; Saied, S. In–Vitro Evaluation and in–Silico Studies Applied on Newly Synthesized Amide Derivatives of N–Phthaloylglycine as Butyrylcholinesterase (BChE) Inhibitors. Comput. Biol. Chem. 2018, 74, 212–217. [Google Scholar] [CrossRef] [PubMed]
  56. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  57. Ellman, G.L.; Courtney, K.D.; Andres, V.; Featherstone, R.M. A New and Rapid Colorimetric Determination of Acetylcholinesterase Activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  58. Pourshojaei, Y.; Abiri, A.; Eskandari, K.; Haghighijoo, Z.; Edraki, N.; Asadipour, A. Phenoxyethyl Piperidine/Morpholine Derivatives as PAS and CAS Inhibitors of Cholinesterases: Insights for Future Drug Design. Sci. Rep. 2019, 9, 19855. [Google Scholar] [CrossRef] [Green Version]
  59. Abd El–Gaber, M.K.; Hassan, H.Y.; Mahfouz, N.M.; Farag, H.H.; Bekhit, A.A. Synthesis, Biological Investigation and Molecular Docking Study of N–Malonyl–1,2–Dihydroisoquinoline Derivatives as Brain Specific and Shelf–Stable MAO Inhibitors. Eur. J. Med. Chem. 2015, 93, 481–491. [Google Scholar] [CrossRef]
Figure 1. Design strategies for innovative compounds of multi–target–directed ligands (MTDLs). (ChE: cholinesterase; MAO: monoamine oxidase; MAO–B: monoamine oxidase B; BuChE: butyrylcholinesterase).
Figure 1. Design strategies for innovative compounds of multi–target–directed ligands (MTDLs). (ChE: cholinesterase; MAO: monoamine oxidase; MAO–B: monoamine oxidase B; BuChE: butyrylcholinesterase).
Molecules 27 09062 g001
Scheme 1. Synthetic routes and required reagents of novel benzothiazole–isoquinoline derivatives.
Scheme 1. Synthetic routes and required reagents of novel benzothiazole–isoquinoline derivatives.
Molecules 27 09062 sch001
Figure 2. The effect of benzothiazole–isoquinoline derivatives (4b, 4c, 4d, 4f, 4g, 4i) on the proliferation of L929 cells (MTT method).
Figure 2. The effect of benzothiazole–isoquinoline derivatives (4b, 4c, 4d, 4f, 4g, 4i) on the proliferation of L929 cells (MTT method).
Molecules 27 09062 g002
Figure 3. (a): Docking model of compound 4g with BuChE. Atomic colors: red carbon atoms of 4g, gray carbon atoms of BuChE residues, blue nitrogen atoms, red oxygen atoms, yellow sulfur atoms. (b): Docking model of compound 4g with MAO–B. Atomic colors: red carbon atoms of 4g, gray carbon atoms of MAO–B residues, blue nitrogen atoms, red oxygen atoms, yellow sulfur atoms.
Figure 3. (a): Docking model of compound 4g with BuChE. Atomic colors: red carbon atoms of 4g, gray carbon atoms of BuChE residues, blue nitrogen atoms, red oxygen atoms, yellow sulfur atoms. (b): Docking model of compound 4g with MAO–B. Atomic colors: red carbon atoms of 4g, gray carbon atoms of MAO–B residues, blue nitrogen atoms, red oxygen atoms, yellow sulfur atoms.
Molecules 27 09062 g003
Table 1. Test results of inhibitory activity of benzothiazole–isoquinoline derivatives 4a4p on MAO.
Table 1. Test results of inhibitory activity of benzothiazole–isoquinoline derivatives 4a4p on MAO.
CompoundsRInhibition Rate of MAO (%)IC50 of MAO (μM)
4ao–Cl14.93>150
4bm–Cl39.2738.82 ± 3.76
4cp–Cl21.5945.90 ± 2.66
4do–Br43.3764.83 ± 4.20
4em–Br9.29128.08 ± 1.27
4fp–Br24.1541.78 ± 2.94
4go–CH357.1114.80 ± 5.45
4hm–CH316.7276.37 ± 2.58
4ip–CH326.4618.53 ± 1.69
4jo–OCH35.70>150
4km–OCH317.7565.48 ± 3.51
4lp–OCH311.3476.32 ± 2.94
4mp–F9.76>150
4np–NO212.51>150
4o2,4–Cl210.1967.80 ± 1.26
Table 2. Test results of inhibitory activity of benzothiazole–isoquinoline derivatives on monoamine oxidase A (MAO–A) and MAO–B.
Table 2. Test results of inhibitory activity of benzothiazole–isoquinoline derivatives on monoamine oxidase A (MAO–A) and MAO–B.
CompoundsRInhibition Rate (%)IC50 (μM)
MAO–AMAO–BCompoundsR
4bm–Cl3.0139.154bm–Cl
4cp–Cl12.0348.454cp–Cl
4do–Br18.0546.004do–Br
4fp–Br15.0446.494fp–Br
4go–CH39.0351.394go–CH3
4ip–CH32.5161.174ip–CH3
Clorgyline84.21Clorgyline
Pargyline93.47Pargyline
Table 3. Test results of inhibitory activity of benzothiazole–isoquinoline derivatives 4a4p on cholinesterase.
Table 3. Test results of inhibitory activity of benzothiazole–isoquinoline derivatives 4a4p on cholinesterase.
CompoundsRInhibition Rate (%)IC50 (μM)
AChEBuChEAChEBuChE
4ao–Cl6.0838.08>150>150
4bm–Cl4.4269.44>15017.59 ± 1.78
4cp–Cl1.1068.80>150>150
4do–Br0.5577.76>15014.61 ± 5.81
4em–Br4.9768.80>15018.27 ± 2.68
4fp–Br1.1032.32>150>150
4go–CH31.1072.64>15030.35 ± 4.59
4hm–CH33.3137.44>15011.52 ± 3.22
4ip–CH32.2147.68>15019.17 ± 2.72
4jo–OCH312.7151.52>150>150
4km–OCH33.3157.92>15010.25 ± 2.91
4lp–OCH33.3141.92>1503.86 ± 0.82
4mp–F2.7622.33>15050.26 ± 1.68
4np–NO22.7620.31>15012.34 ± 3.76
4o2,4–Cl23.8752.16>15021.76 ± 2.85
4p2,4–Br26.6343.84>15027.12 ± 1.73
Tacrine99.4112.43 ± 2.49
Donepezil69.0616.75 ± 1.32
Table 4. Preliminary screening results of antidepressant activity of compounds in the FST.
Table 4. Preliminary screening results of antidepressant activity of compounds in the FST.
CompoundsRAntidepressant Effects
Duration of Immobility (s)DID (%) a
4bm–Cl85.7 ± 8.1 ***51.98
4cp–Cl77.0 ± 18.3 ***56.84
4do–Br62.3 ± 14.0 **65.06
4fp–Br71.7 ± 4.9 ***59.83
4go–CH369.0 ± 15.7 **61.32
4ip–CH383.8 ± 13.6 **53.05
Fluoxetine31.7 ± 7.7 ***82.20
Control178.4 ± 5.9
a DID: percentage decrease in immobility duration. Values are the mean ± SEM (n = 8). Duration of Immobility: * Significantly different compared with control (** p < 0.01, *** p < 0.001).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, W.; Zhao, D.; He, Z.; Hu, Y.; Zhu, Y.; Zhang, L.; Jin, L.; Guan, L.; Wang, S. Synthesis, Characterization and Biological Evaluation of Benzothiazole–Isoquinoline Derivative. Molecules 2022, 27, 9062. https://doi.org/10.3390/molecules27249062

AMA Style

Liu W, Zhao D, He Z, Hu Y, Zhu Y, Zhang L, Jin L, Guan L, Wang S. Synthesis, Characterization and Biological Evaluation of Benzothiazole–Isoquinoline Derivative. Molecules. 2022; 27(24):9062. https://doi.org/10.3390/molecules27249062

Chicago/Turabian Style

Liu, Weihua, Donghai Zhao, Zhiwen He, Yiming Hu, Yuxia Zhu, Lingjian Zhang, Lianhai Jin, Liping Guan, and Sihong Wang. 2022. "Synthesis, Characterization and Biological Evaluation of Benzothiazole–Isoquinoline Derivative" Molecules 27, no. 24: 9062. https://doi.org/10.3390/molecules27249062

Article Metrics

Back to TopTop