Next Article in Journal
Design, Synthesis and In Vitro Mechanistic Investigation of Novel Hexacyclic Cage-Like Hybrid Heterocycles
Previous Article in Journal
X-ray Photospectroscopy and Electronic Studies of Reactor Parameters on Photocatalytic Hydrogenation of Carbon Dioxide by Defect-Laden Indium Oxide Hydroxide Nanorods
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of Novel Triazinoindole-Based Thiourea Hybrid: A Study on α-Glucosidase Inhibitors and Their Molecular Docking

1
Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
2
Neurology Department, King Fahad hospital of University, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 34211, Saudi Arabia
3
Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
4
Department of Chemistry, Hazara University, Mansehra-21300, Khyber Pakhtunkhwa, Pakistan
5
Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor D.E., Malaysia
6
Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor D.E., Malaysia
7
H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
8
Department of Chemistry, Jinnah University for Women, 5-C, Nazimabad, Karachi 74600, Pakistan
*
Authors to whom correspondence should be addressed.
Molecules 2019, 24(21), 3819; https://doi.org/10.3390/molecules24213819
Submission received: 12 September 2019 / Revised: 17 October 2019 / Accepted: 21 October 2019 / Published: 23 October 2019
(This article belongs to the Section Bioorganic Chemistry)

Abstract

:
A new class of triazinoindole-bearing thiosemicarbazides (125) was synthesized and evaluated for α-glucosidase inhibitory potential. All synthesized analogs exhibited excellent inhibitory potential, with IC50 values ranging from 1.30 ± 0.01 to 35.80 ± 0.80 μM when compared to standard acarbose (an IC50 value of 38.60 ± 0.20 μM). Among the series, analogs 1 and 23 were found to be the most potent, with IC50 values of 1.30 ± 0.05 and 1.30 ± 0.01 μM, respectively. The structure–activity relationship (SAR) was mainly based upon bringing about different substituents on the phenyl rings. To confirm the binding interactions, a molecular docking study was performed.

Graphical Abstract

1. Introduction

Diabetes mellitus is a chronic health-threatening metabolic disease that is caused by insufficient insulin secretion and is categorized as hypoglycemia/hyperglycemia [1]. In type II diabetes mellitus, enhanced postprandial glucose levels can increase the risk of developing stroke, atherosclerosis, and other coronary diseases [2]. In order to treat type II diabetes and its complications, the inhibition of digestive enzyme-like α-glucosidase is an effective approach that can reduce postprandial glucose and risk factors [3]: α-glucosidase is located in the epithelium cell lining of the small intestine and is responsible for the conversion of polysaccharides and disaccharides into glucose. The inhibition of α-glucosidase is directly associated with the blood glucose level, and its inhibition is vital due to the potential effect of a decrease in postprandial blood glucose levels [4]. In order to delay rapid blood glucose production, certain α-glucosidase inhibitors such as acarbose and voglibose are used clinically, though there are certain side effect that include abdominal pain, diarrhea, and other gastrointestinal disorders in chronic therapy [5]. Therefore, in order to treat postprandial hyperglycemia, a search for efficient and safe α-glucosidase inhibitors is needed.
Triazinoindole scaffolds possess excellent biological potential against malarial and viral diseases. Substituted triazinoindole scaffolds are of considerable interest due to their excellent antihypertensive [6], antidepressant [7], anti-inflammatory [8], antihypoxic [9], antifungal, and antibacterial activities [10]. Selected triazinoindole compounds act as potential drugs in treating the common cold [11,12,13,14].
Our research group has been working on the design and synthesis of heterocyclic compounds in search of potential lead compounds for many years, and we have found promising results [15,16,17,18,19,20,21,22,23,24,25,26,27,28,29]. We have already reported on triazinoindole analogs as potent α-glucosidase inhibitors [30]. Thus, we decided to screen a library of triazinoindole-bearing thiosemicarbazide analogs for α-glucosidase activity (Figure 1).

2. Results and Discussion

2.1. Chemistry

A new class of triazinoindole-based thiosemicarbazide analogs (125) was carried out in three steps.
In the first step, thiosemicarbazide was reacted and refluxed with isatin in H2O in the presence of potassium carbonate to yield 5H-triazinoindole-3-thiol as an intermediate product (I). The intermediate (I) was then mixed and refluxed with different substituted phenacyl bromides in EtOH in the presence of Et3N to give triazinoindole derivatives as a second intermediate product (II). (Scheme 1)
In the second step, hydrazine hydrate was reacted and refluxed with different isothiocyanates in methanol to yield a thiosemicarbazide derivative as an intermediate (III). (Scheme 2)
In the third step, intermediate product (II) was reacted and refluxed with intermediate product (III) in glacial acetic acid to give the final product, triazinoindole-bearing thiosemicarbazide (125). (Scheme 3, Table 1).

2.2. Biological Activity

A new class of triazinoindole-bearing thiosemicarbazide analogs (125) was synthesized and was evaluated for α-glucosidase inhibitory potential. All of the synthesized scaffolds exhibited outstanding inhibitory potential, with IC50 values ranging from 1.30 ± 0.01 to 35.80 ± 0.80 μM when compared to standard acarbose with an IC50 value of 38.60 ± 0.20 μM. The structure–activity relationship (SAR) was mainly based upon different substation pattern on phenyl rings.
We compared compound 1 (IC50 = 1.30 ± 0.05 μM) with a methoxy moiety at position 4 on one phenyl ring and two chloro groups at positions 2 and 3 on the second phenyl ring to scaffold 18 (IC50 = 2.30 ± 0.05 μM), which also had a methoxy moiety at position 4 on one phenyl ring and two chloro groups at positions 3 and 4 on the second phenyl ring. The inhibition difference in these two scaffolds may have been due to two chloro groups that were present in different positions on the second phenyl ring.
If we compare scaffold 9 (IC50 value 6.80 ± 0.10 μM) to scaffold 17 (IC50 value 8.80 ± 0.20 μM), both scaffolds had a nitro moiety on one phenyl ring, and in both cases the other phenyl ring was unsubstituted. In scaffold 9, the nitro moiety was present at position 4, while in scaffold 17, the nitro moiety was present at position 3 on the phenyl ring. The potential difference in these two scaffolds may have been due to the nitro moiety being in a different position on one phenyl ring.
Similarly, we compared compound 15 (IC50 = 5.80 ± 0.20 μM) with a methoxy moiety at position 4 on one phenyl ring and two methyl groups at positions 2 and 6 on the second phenyl ring to scaffold 22 (IC50 = 5.90 ± 0.10 μM), which also had a methoxy moiety at position 4 on one phenyl ring and two methyl groups at positions 2 and 3 on the second phenyl ring. The inhibition difference in these two scaffolds may have been due to two methyl groups that were present in different positions on the second phenyl ring.
It was observed over the whole study that the phenyl ring substituents’ nature as well as their positions greatly affected the inhibitory potential of the compound. A docking study was done to understand the binding interactions of the most active scaffolds with the enzyme active site.

2.3. Docking Study

Docking studies were carried out on scaffolds 1, 16, and 23, which displayed the most potent inhibitory potential among the whole series. Prior to docking and analysis of the binding mode of the most active compound, 1, the docking method was validated through the control docking of a native inhibitor. Acarbose was docked into α-glucosidase from sugar beet (PDB code: 3W37) and was compared by superimposing the native ligand in the protein, as mentioned in Imran et. al. (2016) [24]. Prior to docking and analysis of the binding mode of the active compounds, the docking method was validated through the control docking of a native inhibitor. Acarbose was docked into α-glucosidase from sugar beet (PDB code: 3W37) and was compared by superimposing the native ligand in the protein (Figure 2a). The rmsd value between the docked and actual pose of acarbose was found to be 0.65 Å. Even though α-glucosidase from sugar beet shared a relatively low homology with Baker’s yeast α-glucosidase (16% identity), the active site was highly conserved, and the main interactions of the ligand remained the same. Another control docking was done on the target protein, isomaltase from Baker’s yeast, using α-d-glucopyranose, which was the native ligand located in the active site (Figure 2b). The rmsd value for the docked pose and the native ligand was found to be 0.93 Å.
The Chemscore values of the active compounds are reported in Table 2, and these values correlated well with the IC50 values. The results obtained showed that scaffold 1 recorded the highest Chemscore value, −89.3 kJ/mol, followed by compound 23, which recorded a Chemscore value of −87.7 kJ/mol, and finally compound 16, which displayed the lowest Chemscore value, −74.5.
The results from the docking of these active compounds showed that they were able to form several hydrogen bonds within the cavity. The hydrogen on the nitrogen of triazinoindole formed a hydrogen bond with the backbone (Oε2) of Glu276, and the catalytic residue was involved in the hydrolysis reaction at a distance of 2.16 Å.
In the case of compound 1, one of the nitrogens on the triazine moiety established a hydrogen bonding interaction with the residue of hydrophobic patch Phe300 at a distance of 3.42 Å. The sulfur linkage displayed hydrogen bonding with the side chain (O) of Glu304 at a distance of 2.23 Å. An interaction involving a halide bond was observed between a chlorine substituent at the meta position and the side chain (O) of Thr307 at a distance of 1.89 Å. An aromatic ring containing a methoxy substituent formed an electrostatic π-hydrogen interaction with His239 (Hε1) at a distance of 2.64 Å. As for the triazine moiety, an electrostatic π-hydrogen involving the residue of hydrophobic patch Phe177 was expected to stabilize the ligand–enzyme complex alongside Tyr71, which formed an electrostatic π-hydrogen interaction with one of the hydrogens from the triazinoindole moiety at a distance of 3.93 Å Figure 3.
The analog 23 docking study revealed that this scaffold was capable of forming several hydrogen bonds within the cavity. The residue Glu276, through its backbone (Oε2), participated in hydrogen bonding interactions with the amino group of triazinoindole compound 23 at a distance of 2.43 Å. As for the triazine moiety, electrostatic π-hydrogen was involved in the side chain of residue Phe157, which was expected to stabilize the ligand–enzyme complex. Another hydrogen formed an electrostatic π-hydrogen interaction with Tyr71. It was also observed that His239 was capable of forming an electrostatic interaction with the oxygen of the nitro substituent at the meta position. An aromatic ring consisting of dichloro substituents were stabilized through electrostatic interactions with Pro309 through electrostatic π-hydrogen interactions (Figure 4).
The docking results for compound 16 displayed fewer interactions compared to compounds 1 and 23. Some of the interactions that remained the same were the interactions of hydrogen on the nitrogen of triazinoindole, which formed a hydrogen bond with the backbone (Oε2) of Glu276 at a distance of 1.84 Å, and hydrogen formed electrostatic π-hydrogen interactions with Tyr71 at a distance of 3.98 Å. On the other hand, an aromatic ring consisting of a fluoro substituent at the para position displayed an electrostatic interaction with Glu304. It was observed that the CH3 of methoxy at the para position of the other aromatic ring displayed π-hydrogen interactions with Phe157 at a distance of 4.12 Å. (Figure 5)

3. Conclusions

In conclusion, we synthesized 25 analogs of triazinoindole-bearing thiosemicarbazide and evaluated them against the α-glucosidase enzyme. All of the synthesized scaffolds exhibited outstanding inhibitory potential, with IC50 values ranging from 1.30 ± 0.01 to 35.80 ± 0.80 μM when compared to standard acarbose with an IC50 value of 38.60 ± 0.20 μM. It was confirmed through the SAR that polar- and electron-withdrawing groups on the phenyl rings had a lot of influence on the potency of the compounds. A docking study was done to understand the binding interactions of the most active scaffolds.

4. Experiment

4.1. General Method for the Synthesis of Triazinoindole-Bearing Thiosemicarbazide Analogs (125)

A new class of triazinoindole-bearing thiosemicarbazide analogs (125) was carried out in three steps.
In the first step, thiosemicarbazide (10 mmol) was reacted and refluxed with isatin (10 mmol) in H2O in the presence of potassium carbonate (5 mmol) to yield 5H-triazinoindole-3-thiol as intermediate (I). Intermediate product (I) (5 mmol) was then mixed and refluxed with different substituted phenacyl bromides (5 mmol) in EtOH in the presence of Et3N to give a triazinoindole derivative as the second intermediate product (II).
In the second step, hydrazine hydrate (2 mL) was reacted and refluxed with different isothiocyanates (1 mmol) in methanol to yield a thiosemicarbazide derivative as intermediate product (III).
In the third step, the intermediate (II) was reacted and refluxed with an equimolar intermediate (III) in glacial acetic acid to give the final product, triazinoindole-bearing thiosemicarbazide.
2-(2-((5H-[1,2,4]triazino [5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(2,3-dichlorophenyl)hydrazine-1-carbothioamide (1) Yield: 62%; 1H-NMR: (500 MHz, DMSO-d6), δ 13.30 (s, 1H, NH), 11.25 (s, 2H, NH), 7.84 (d, J = 7.3 Hz, 2H, Ar), 7.54 (d, J = 6.2 Hz, 1H, Ar), 7.45 (s, 1H, Ar), 7.35 (t, J = 5 Hz, 2H, Ar), 7.26 (dd, J = 1, 6.65 Hz, 1H, Ar), 7.10 (m, 4H, Ar), 5.2 (s, 2H, CH2), 3.81 (s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 159, 150.9, 141.2, 131.8, 131.8, 130.3, 128.2, 127.0, 126.9, 126.7, 123, 122.3, 121.2, 119.7, 119.7, 118.3 114.1, 114.1, 111, 104, 55.2, 41.2. HREI-MS: m/z, calcd for C25H19Cl2N7OS2 [M]+ 567.0470; found: 567.0458.
2-(2-((5H-[1,2,4]triazino [5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(4-nitrophenyl)hydrazine-1-carbothioamide (2) Yield: 68%; 1H-NMR: (500 MHz, DMSO-d6), δ 11.25 (s, 1H, NH), 10.86 (s, 2H, NH), 8.24 (d, J = 7.3 Hz, 2H, Ar), 7.83 (d, J = 7.2 Hz, 2H, Ar), 7.79 (d, J = 7.65 Hz, 2H, Ar), 7.53 (d, J = 6.2 Hz, 1H, Ar), 7.34 (d, J = 1H, Ar), 7.10 (t, J = 6.3, 1H, Ar), 6.98 (t, J = 7.5, 3H, Ar), 4.91 (s, 2H, CH2), 3.81 (s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6), δ 171.9, 165.8, 163.1, 159, 150.9, 141.2, 131.8, 131.8, 130.3, 128.2, 127, 126.9, 126.7, 123, 122.3, 121.2, 119.7, 119.7, 118.3 114.1, 114.1, 111, 104, 55.2, 38.2. HREI-MS: m/z, calcd for C25H20N8O3S2 [M]+ 544.1100; found: 544.1088.
2-(2-((5H-[1,2,4]triazino [5,6-b]indol-3-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene)-N-(2,3-dichlorophenyl)hydrazine-1-carbothioamide (3) Yield: 68%; 1H-NMR: (500 MHz, DMSO-d6), δ 11.25 (s, 1H, NH), 10.86 (s, 2H, NH), 8.24 (d, J = 7.3 Hz, 2H, Ar), 7.83 (d, J = 7.2 Hz, 2H, Ar), 7.79 (d, J = 7.65 Hz, 2H, Ar), 7.53 (d, J = 6.2 Hz, 1H, Ar), 7.34 (d, J = 6.5 Hz, 1H, Ar), 7.10 (t, J = 6.3 Hz, 1H, Ar), 6.98–6.93 (m, 3H, Ar), 4.91 (s, 2H, CH2), 3.81 (s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6), δ 171.9, 165.8, 163.1, 159, 150.9, 141.2, 131.8, 131.8, 130.3, 128.2, 127, 126.9, 126.7, 123, 122.3, 121.2, 119.7, 119.7, 118.3 114.1, 114.1, 111, 104, 55.2, 38.2. HREI-MS: m/z, calcd for C25H20N8O3S2 [M]+ 544.1100; found: 544.1088.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene)-N-(p-tolyl)hydrazine-1-carbothioamide (4) Yield: 72%; 1H-NMR: (500 MHz, DMSO-d6), δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 8.2 (d, J = 7 Hz, 1H, Ar), 8.01 (m, 2H, Ar), 7.9 (d, J = 7 Hz, 1H, Ar), 7.80 (d, J = 6.2 Hz, 1H, Ar), 7.7 (m, 5H, Ar), 7.6 (d, J = 7.1 Hz, 2H, Ar), 7.4 (m, 3H, Ar), 7.1 (t, J = 6.25 Hz, 1H, Ar), 6.9 (d, J = 6.5 Hz, 1H, Ar), 4.9 (s, 2H, CH2), 3.0 (s, 3H, CH3). 13C-NMR (125 MHz, DMSO-d6), δ 192.8, 166.2, 163.1, 146.4, 144.8, 141, 139.5, 139.4, 138.8, 134.7, 133, 129.0, 129.0, 128.9, 128.4, 127.5, 127.0, 126.8, 126.8, 126.4, 126.2,122.4, 122.3, 121.4, 119.8, 117.5, 112.6, 111.0, 106.9, 38.2, 21.3. HREI-MS: m/z, calcd for C31H25N7S2 [M]+ 559.1613; found: 559.1601.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(p-tolyl)hydrazine-1-carbothioamide (5) Yield: 73%; 1H-NMR: (500 MHz, DMSO-d6), δ 13.33 (s, 1H, NH), 11.25 (s,2H, NH), 7.8 (d, J = 7.25 Hz, 2H, Ar), 7.79 (d, J = 7.2 Hz, 2H, Ar), 7.54 (d, J = 6.2 Hz, 2H, Ar), 7.35 (m, 2H, Ar), 7.11 (t, J = 6.25 Hz, 2H, Ar), 7.02 (m, 2H, Ar), 5.2 (s, 2H, CH2), 3.80 (s, 3H, -OCH3), 2.42 (s, 3H, -CH3). 13C-NMR (125 MHz, DMSO-d6), δ 165.8, 163.1, 159.0, 150.9, 141.2, 131.8, 130.3, 127.0, 127.0, 127.0, 126.9, 126.7, 122.3, 122.3, 121.0, 119.7, 119.7,114.1, 114.1, 114.1, 114.0, 111.0,104.6, 55.2, 38.2, 22.0. HREI-MS: m/z, calcd for C26H23N7OS2 [M]+ 513.1405; found: 513.1390.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene)-N-(2-bromophenyl)hydrazine-1-carbothioamide (6) Yield: 77%; 1H-NMR: (500 MHz, DMSO-d6), δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 8.0 (d, J = 7 Hz, 1H, Ar), 7.9 (d, J = 7 Hz, 1H, Ar), 7.7 (d, J = 6.2 Hz, 1H, Ar), 7.5 (d, J = 7 Hz, 2H, Ar), 7.4 (d, J = 7.1 Hz, 2H, Ar), 7.36 (m, 3H, Ar), 7.34 (t, J = 6.25 Hz, 2H, Ar), 7.24 (d, J = 6.6 Hz, 1H, Ar), 7.21 (t, J = 7.3 Hz, 1H, Ar), 7.17 (d, J = 7 Hz, 1H, Ar), 7 (t, J = 6.9 Hz, 1H, Ar), 6.9 (d, J = 6.5 Hz, 1H, Ar), 4.9 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6), δ 192.8, 166.2, 163.1, 146.4, 144.8, 141, 139.5, 139.4, 138.8, 134.7, 133, 129.0, 129.0, 128.9, 128.4, 127.5, 127.0, 126.8, 126.8, 126.4, 126.2,122.4, 122.3, 121.4, 119.8, 117.5, 112.6, 111.0, 106.9, 38.2. HREI-MS: m/z, calcd for C30H22BrN7S2 [M]+ 623.0561; found: 623.0550.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(p-tolyl)ethylidene)-N-(2-bromophenyl)hydrazine-1-carbothioamide (7) Yield: 68%; 1H-NMR: (500 MHz, DMSO-d6), δ 12.5 (s, 2H, NH), 11.25 (s, 1H, NH), 8.28 (d, J = 6.45 Hz, 1H, Ar), 8.01 (d, J = 6.75 Hz, 3H, Ar), 7.69 (t, J = 6.65 Hz, 2H, Ar), 7.5 (d, J = 6.97 Hz, 2H, Ar), 7.43 (m, 4H, Ar), 4.8 (s, 2H, CH2), 2.42 (s, 3H, CH3). 13C-NMR (125 MHz, DMSO-d6), δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.2, 125.6, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0, 109.7, 105.8, 38.2, 20.7. HREI-MS: m/z, calcd for C25H20BrN7S2 [M]+ 561.0405; found: 561.0392.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(p-tolyl)ethylidene)-N-(p-tolyl)hydrazine-1-carbothioamide (8) Yield: 65%; 1H-NMR: (500 MHz, DMSO-d6): δ 12.5 (s, 2H, NH), 11.4 (s, 1H, NH), 8.25 (d, J = 6.45 Hz, 1H, Ar), 7.98 (d, J = 6.75 Hz, 3H, Ar), 7.69 (m, 2H, Ar), 7.48 (d, J = 6.97 Hz, 2H, Ar), 7.39 (m, 4H, Ar), 4.9 (s, 2H, CH2), 2.42 (s, 6H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0, 109.7, 105.8, 38.2, 20.7, 20.7. HREI-MS: m/z, calcd for C26H23N7S2 [M]+ 497.1456; found: 497.1440.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-nitrophenyl)ethylidene)-N-phenylhydrazine-1-carbothioamide (9) Yield: 71%; 1H-NMR: (500 MHz, DMSO-d6): δ 13.34 (s, 1H, NH), 11.25 (s, 2H, NH), 7.79 (d, J = 6.75 Hz, 3H, Ar), 7.54 (d, J = 5.8 Hz, 3H, Ar), 7.36 (ddd, J = 0.9, 6.4 Hz, 1H, Ar), 7.28 (dd, J = 6.55 Hz, 3H, Ar), 7.11 (m, 1H, Ar), 6.97 (d, J = 6.5 Hz, 2H, Ar), 5.1 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0 105.8, 38.2. HREI-MS: m/z, calcd for C24H18N8O2S2 [M]+ 514.0994; found: 514.0980.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(2-bromophenyl)hydrazine-1-carbothioamide (10) Yield: 71%; 1H-NMR: (500 MHz, DMSO-d6): δ 13.30 (s, 1H, NH), 11.25 (s, 1H, NH), 10.86 (s, 1H, NH), 7.84 (t, J = 7.2 Hz, 2H, Ar), 7.54 (d, J = 6.2 Hz, 2H, Ar), 7.35 (t, J = 6.35, 2H, Ar), 7.10 (t, J = 6.55, 2H, Ar), 7.03 (d, J = 7.1 Hz, 2H, Ar), 6.99 (m, 2H, Ar), 4.80 (s, 2H, CH2), 3.81 (s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.2, 125.6, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0, 109.7, 105.8, 38.2, 55.2. HREI-MS: m/z, calcd for C25H20BrN7OS2 [M]+ 577.0354; found: 577.0342.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(3-nitrophenyl)ethylidene)-N-(2-bromophenyl)hydrazine-1-carbothioamide (11) Yield: 70%; 1H-NMR: (500 MHz, DMSO-d6): δ 13.30 (s, 1H, NH), 11.25 (s, 2H, NH), 8.71 (m, 2H, Ar), 8.37 (d, J = 6.5 Hz, 1H, Ar), 8.33 (d, J = 6.35, 1H, Ar), 8.21 (m, 2H, Ar), 7.95 (s, 1H, Ar), 7.75 (t, J = 6.6 Hz, 1H, Ar), 7.55 (d, J = 6.25 Hz, 1H, Ar), 7.36 (t, J = 6.15 Hz, 1H, Ar), 7.11 (t, J = 6.2, 1H, Ar), 6.9 (d, J = 6.5, 1H, Ar), 5.5 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.4, 123.3, 121.5, 119.7, 119.7, 111.0 105.8, 38.2. HREI-MS: m/z, calcd for C24H17BrN8O2S2 [M]+ 592.0099; found: 592.0084.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(3-nitrophenyl)ethylidene)-N-(p-tolyl)hydrazine-1-carbothioamide (12) Yield: 70%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 7.80 (m, 3H, Ar), 7.68 (s, 1H, Ar), 7.54 (d, J = 4.8 Hz, 2H, Ar), 7.35 (t, J = 6.4, 1H, Ar), 7.25 (m, 3H, Ar), 7.11 (t, J = 6.3, 1H, Ar), 6.98 (m, 1H, Ar), 4.8 (s, 2H, CH2), 1.9 (s, 3H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.4, 122.3, 121, 119.7, 119.7, 111.0 105.8, 38.2, 21.0. HREI-MS: m/z, calcd for C25H20N8O2S2 [M]+ 528.1151; found: 528.1136.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene)-N-(2,6-dimethylphenyl)hydrazine-1-carbothioamide (13) Yield: 64%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 8.0 (d, J = 7 Hz, 1H, Ar), 7.9 (d, J = 7 Hz, 1H, Ar), 7.7 (d, J = 6.2 Hz, 1H, Ar), 7.5 (d, J = 7, 2H, Ar), 7.4 (d, J = 7.1 Hz, 2H, Ar), 7.36 (m, 3H, Ar), 7.34 (d, J = 6.25 Hz, 2H, Ar), 7.21 (d, J = 7.3 Hz, 1H, Ar), 7.17 (d, J = 7 Hz, 1H, Ar), 7 (t, J = 6.9 Hz, 1H, Ar), 6.9 (d, J = 6.5 Hz, 1H, Ar), 4.9 (s, 2H, CH2), 1.88 (s, 6H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 192.8, 166.2, 163.1, 146.4, 144.8, 141, 139.5, 139.4, 138.8, 134.7, 133.5, 129.0, 129.0, 128.9, 128.4, 127.5, 127.0, 126.9, 126.8, 126.4, 126.2,122.4, 122.3, 121.4, 119.8, 117.5, 112.6, 111.0, 106.9, 38.2, 21.0, 21.0. HREI-MS: m/z, calcd for C32H27N7S2 [M]+ 573.1769; found: 573.1755.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(3-nitrophenyl)ethylidene)-N-(2,6-dimethylphenyl)hydrazine-1-carbothioamide (14) Yield: 70%; 1H-NMR: (500 MHz, DMSO-d6): δ 12.57 (s, 2H, NH), 11.28 (s, 1H, NH), 8.36 (t, J = 6.5 Hz, 1H, Ar), 8.6(s, 1H, Ar), 8.28 (d, J = 6.4, 2H, Ar), 7.96 (m, 3H, Ar), 7.56 (d, J = 6.75, 2H, Ar), 7.43 (m, 2H, Ar), 5(s, 2H, CH2), 1.91 (s, 6H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.4, 122.3, 121, 119.7, 119.7, 111.0, 105.8, 38.2, 22.1 22.1. HREI-MS: m/z, calcd for C26H22N8O2S2 [M]+ 542.1307; found: 542.1293.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(2,6-dimethylphenyl)hydrazine-1-carbothioamide (15) Yield: 75%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 7.84 (d, J = 7.25 Hz, 3H, Ar), 7.54 (d, J = 6.2 Hz, 1H, Ar), 7.34 (d, J = 6.5 Hz, 1H, Ar), 7.10 (t, J = 6.3, 1H, Ar), 6.99 (t, J = 7.2 Hz, 5H, Ar), 4.4 (s, 2H, CH2), 3.81 (s, 3H, OCH3), 2.3 (s, 6H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 165.8, 163.1, 159.0, 150.9, 141.2, 131.8, 130.3, 127.0, 127.0, 127.0, 126.9, 126.7, 122.3, 122.3, 121.0, 119.7, 119.7,114.1, 114.1, 114.1, 114.0, 111.0,104.6, 55.2, 38.2, 22.2, 22.2. HREI-MS: m/z, calcd for C27H25N7OS2 [M]+ 527.1562; found: 527.1551.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(4-fluorophenyl)hydrazine-1-carbothioamide (16) Yield: 74%; 1H-NMR: (500 MHz, DMSO-d6): δ 12.5 (s, 2H, NH), 10.86 (s, 1H, NH), 8.28 (d, J = 6.5 Hz, 1H, Ar), 7.90 (d, J = 7.3 Hz, 2H, Ar), 7.66 (m, 1H, Ar), 7.55 (d, J = 6.75 Hz, 1H, Ar), 7.43 (d, J = 6.4 Hz, 2H, Ar), 7.39 (m, 1H, Ar), 7.08 (d, J = 7.3 Hz, 2H, Ar), 7.02 (d, J = 6.75 Hz, 2H, Ar), 4.94 (s, 2H, CH2), 3.46 (s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6): δ 191.5, 166.3, 163.4, 163.3 146.7, 146.3, 140.9, 140.2, 131.5, 131.1, 130.8, 130.6, 129.2, 128.7, 128.6, 122.4, 122.2, 121.7, 121.3, 117.5, 114.0, 113.9, 112.6, 55.5, 38.0. HREI-MS: m/z, calcd for C25H20FN7OS2 [M]+ 517.1155; found: 517.1143.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(3-nitrophenyl)ethylidene)-N-phenylhydrazine-1-carbothioamide (17) Yield: 69%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.27 (s, 2H, NH), 10.86 (s, 1H, NH), 8.6 (s, 1H, Ar), 8.36 (d, J = 6.5, 3H, Ar), 8.19 (m, 1H, Ar), 7.9(d, J = 5.2, 1H, Ar), 7.75 (m, 2H, Ar), 7.55 (d, J = 6.15 Hz, 2H, Ar), 7.36 (t, J = 6.35, 1H, Ar), 7.18 (t, J = 6.3, 1H, Ar), 6.98 (d, J = 6.4, 1H, Ar), 4.9 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 130.4, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0, 105.8, 38.2. HREI-MS: m/z, calcd for C24H18N8O2S2 [M]+ 514.0994; found: 514.0980.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl) ethylidene)-N-(3,4-dichlorophenyl)hydrazine-1-carbothioamide (18) Yield: 71%; 1H-NMR: (500 MHz, DMSO-d6): δ 12.6 (s, 2H, NH), 11.25 (s, 1H, NH), 8.28 (d, J = 6.45 Hz, 1H, Ar), 8.10 (d, J = 7.3 Hz, 2H, Ar), 7.69 (m, 2H, Ar), 7.61 (s, 1H, Ar), 7.55 (d, J = 6.7 Hz, 1H, Ar), 7.43 (t, J = 6.3 Hz, 2H, Ar), 7.12 (d, J = 7.35 Hz, 2H, Ar), 4.9 (s, 2H, CH2), 3.81(s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 159, 150.9, 141.2, 131.8, 131.8, 130.3, 128.2, 127, 126.9, 126.7, 123, 121.3, 121.2, 119.7, 119.7, 118.3 114.1, 114, 111, 104, 55.2, 38.2. HREI-MS: m/z, calcd for C25H19Cl2N7OS2 [M]+ 567.0470; found: 567.0458.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene)-N-(3,4-dichlorophenyl)hydrazine-1-carbothioamide (19) Yield: 77%; 1H-NMR: (500 MHz, DMSO-d6): δ 12.5 (s, 2H, NH), 11.3 (s, 1H, NH), 8.27 (d, J = 6.5 Hz, 1H, Ar), 8.21 (d, J = 6.95 Hz, 2H, Ar), 7.91 (d, J = 6.9 Hz, 2H, Ar), 7.80 (d, J = 6.2 Hz, 2H, Ar), 7.69 (d, J = 7Hz, 2H, Ar), 7.55 (m, 4H, Ar), 7.47(m, 3H, Ar), 5.03 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6): δ 192.8, 166.2, 163.1, 146.4, 144.8, 141, 139.5, 139.4, 138.8, 134.7, 133, 129.0, 129.0, 128.9, 128.4, 127.5, 127.0, 126.9, 126.8, 126.4, 126.2,122.6, 122.3, 121.4, 119.8, 117.5, 112.6, 111.0, 106.9, 38.2. HREI-MS: m/z, calcd for C30H21Cl2N7S2 [M]+ 613.0677; found: 613.0664.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(p-tolyl)ethylidene)-N-(2,6-dimethylphenyl)hydrazine-1-carbothioamide (20) Yield: 68%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 7.79 (d, J = 6.6 Hz, 2H, Ar), 7.53 (d, J = 4.1 Hz, 3H, Ar), 7.35 (t, J = 6.45 Hz, 1H, Ar), 7.23 (d, J = 6.5 Hz, 3H, Ar), 7.10 (t, J = 6.3 Hz, 1H, Ar), 6.97 (d, J = 6.5, 1H, Ar), 2.3(s, 6H, CH3), 4.8 (s, 2H, CH2), 1.91 (s, 3H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 129.3, 129.3, 129.2, 129.2, 125.6, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0, 109.7, 105.8, 38.2, 21.0, 21.0, 20.7. HREI-MS: m/z, calcd for C27H25N7S2 [M]+ 511.1613; found: 511.1600.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(p-tolyl)ethylidene)-N-(3,4-dichlorophenyl)hydrazine-1-carbothioamide (21) Yield: 68%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 7.77 (d, J = 6.4 Hz, 2H, Ar), 7.54 (d, J = 4.8 Hz, 2H, Ar), 7.32 (t, J = 6.2 Hz, 1H, Ar), 7.23 (d, J = 6.5 Hz, 3H, Ar), 7.10 (t, J = 6.25 Hz, 2H, Ar), 6.97 (s, 1H, Ar), 4.9 (s, 2H, CH2), 1.91 (s, 3H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 159, 150.9, 141.2,131.8, 131.8, 130.3, 128.2, 127, 126.9, 126.7, 123, 121.3, 121.2, 119.7, 119.7, 118.3 114.1, 114, 111, 104, 38.2, 20.1. HREI-MS: m/z, calcd for C25H19Cl2N7S2 [M]+ 551.0520; found: 551.0510.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-(2,3-dimethylphenyl)hydrazine-1-carbothioamide (22) Yield: 62%; 1H-NMR: (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.86 (s, 1H, NH), 7.79 (d, J = 6.6 Hz, 2H, Ar), 7.51 (d, J = 4.5 Hz, 3H, Ar), 7.30 (t, J = 6.45 Hz, 1H, Ar), 7.20 (d, J = 6.7 Hz, 3H, Ar), 7.10 (t, J = 6.3 Hz, 1H, Ar), 6.93 (d, J = 6.5, 1H, Ar), 4.8 (s, 2H, CH2), 3.8 (s, 3H, OCH3), 2.3 (s, 6H, CH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 151.1, 141.2, 137.2, 131.9, 131.2, 130.4, 129.3, 129.3, 129.3, 129.2, 125.6, 125.6, 125.4, 122.3, 121.2, 119.7, 119.7, 111.0, 109.7, 105.8, 55.2, 38.2, 21.0, 21.0. HREI-MS: m/z, calcd for C27H25N7OS2 [M]+ 527.1562; found: 527.1551.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(3-nitrophenyl)ethylidene)-N-(2,3-dichlorophenyl)hydrazine-1-carbothioamide (23) Yield: 65%; 1H-NMR (500 MHz, DMSO-d6): δ 11.29 (s, 2H, NH), 10.86 (s, 1H, NH), 8.72 (d, J = 6.4 Hz, 1H, Ar), 8.37 (m, 1H, Ar), 8.23 (d, J = 6.2, 1H, Ar), 7.96 (s, 1H, Ar), 7.75 (m, 3H, Ar), 7.56 (d, J = 6.25, 1H, Ar), 7.37 (t, J = 6.25, 1H, Ar), 7.12 (t, J = 6.25, 1H, Ar), 6.98 (d, J = 6.5, 1H, Ar), 4.8 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 159, 150.9, 141.2, 131.8, 131.8, 130.3, 129.2, 127, 126.9, 126.7, 123, 122.3, 121.2, 119.7, 119.7, 118.3 114.1, 114, 111, 104, 38.2. HREI-MS: m/z, calcd for C24H16Cl2N8O2S2 [M]+ 582.0215; found: 582.0202.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene)-N-phenylhydrazine-1-carbothioamide (24) Yield: 74%; 1H-NMR (500 MHz, DMSO-d6): δ 11.27 (s, 2H, NH), 10.86 (s, 1H, NH), 8.2 (d, J = 6.8 Hz, 2H, Ar), 7.75 (m, 7H, Ar), 7.55 (d, J = 6.2 Hz, 1H, Ar), 7.49 (t, J = 6.3 Hz, 3H, Ar), 7.39 (m, 3H, Ar), 7.19 (t, J = 6.25 Hz, 1H, Ar), 6.98 (d, J = 6.45 Hz, 1H, Ar), 5.03 (s, 2H, CH2). 13C-NMR (125 MHz, DMSO-d6): δ 192.8, 166.2, 163.1, 146.4, 144.8, 141, 139.5, 139.4, 138.8, 134.7, 133, 129.0, 129.0, 128.9, 128.4, 127.5, 127.0, 126.8, 126.8, 126.4, 126.2, 122.4, 122.3, 121.4, 119.8, 117.5, 112.6, 111.0, 106.9, 38.2. HREI-MS: m/z, calcd for C30H23N7S2 [M]+ 545.1456; found: 545.1439.
2-(2-((5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio)-1-(4-methoxyphenyl)ethylidene)-N-phenylhydrazine-1-carbothioamide (25) Yield: 73%; 1H-NMR (500 MHz, DMSO-d6): δ 11.25 (s, 2H, NH), 10.6 (s, 1H, NH), 7.84 (d, J = 7.3 Hz, 3H, Ar), 7.54 (d, J = 6.25 Hz, 1H, Ar) 7.4 (m, 1H, Ar), 7.36 (t, J = 6.4 Hz, 1H, Ar), 7.11 (t, J = 6.75 Hz, 2H, Ar), 6.99 (t, J = 7.1 Hz, 5H, Ar), 4.9 (s, 2H, CH2), 3.84 (s, 3H, OCH3). 13C-NMR (125 MHz, DMSO-d6): δ 171.9, 165.8, 163.1, 159.0,150.9, 141.2, 131.8, 130.4, 127.0, 126.9, 126.9, 126.9, 126.7, 122.3, 121.2, 119.7, 119.7,114.2, 114.0, 114.0, 114.0, 111.0, 104.6, 55.2, 38.2. HREI-MS: m/z, calcd for C25H21N7OS2 [M]+ 499.1249; found: 499.1233.

4.2. α-Glucosidase Assay Protocol

The α-glucosidase activity was executed according to Fazal et al. [31]. The following chemicals were used (concentrations):
  • 70 μL of 50 mM phosphate buffer (pH 6.8);
  • 10 μL (0.5 mM in methanol) test compounds; and
  • 10 μL (0.057 units, Sigma Inc.) of enzyme solution in buffer.
For details on the experiment, kindly see Reference [31].

4.3. Molecular Docking

Molecular docking was performed on the active compounds to identify possible binding modes that explained the reason for their potency. The method used for molecular docking was as mentioned in our previous paper with slight modifications [32]. The molecular docking study was conducted using a homology model for α-glucosidase. The structures of all compounds were prepared using Chem3D by CambridgeSoft. The geometry and energy of the structures were optimized using MMFF94. GOLD was used to identify the binding modes of the active compounds responsible for the activity. The Chemscore fitness function with default settings was employed in this study. The protein sequence for Baker’s yeast α-glucosidase (MAL12) was obtained from uniprot (http://www.uniprot.org). A homology model for Saccharomyces. cerevisiae glucosidase was built using the crystal structure of α-d-glucose-bound isomaltase from S. cerevisiae (PDB ID: 3A4A), which shares a 72% identical and 85% similar sequence to α-glucosidase. The sequence alignment and homology modeling were performed using Swiss-Model, which is a fully automated homology modeling pipeline (SWISS-MODEL) managed by the Swiss Institute of Bioinformatics. The docking results were visualized using Discovery Studio visualizer 3.5 and PyMol. The homology model was evaluated using PROCHECK.

Author Contributions

Conceptualization, M.T. and F.J.A.; methodology, F.R.; software, S.I.; validation, S.I., and F.N.; formal analysis, H.U.; investigation, S.H.; resources, K.Z.; data curation, M.T. and F.R.; writing—original draft preparation, X.X.; writing—review and editing, X.X.; visualization, X.X.; supervision, K.M.K. project administration, F.J.A., F.R. and; funding acquisition, F.J.A.

Funding

The authors would like to acknowledge the Higher Education Commission of Pakistan for providing a research grant under the National Research Program for Universities (Project Nos. 5721 & 5092).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fatmawati, S.; Shimizu, K.; Kondo, R. Ganoderol B: A potent α-glucosidase inhibitor isolated from the fruiting body of Ganoderma lucidum. Phytomedicine 2011, 18, 1053. [Google Scholar] [CrossRef]
  2. Rother, K.I. Diabetes treatment—Bridging the divide. New Engl. J. Med. 2007, 356, 1499. [Google Scholar] [CrossRef] [PubMed]
  3. Casirola, D.M.; Ferraris, R.P. Alpha-Glucosidase inhibitors prevent diet-induced increases in intestinal sugar transport in diabetic mice. Metabolism 2006, 55, 832–841. [Google Scholar] [CrossRef] [PubMed]
  4. Chiasson, J.L.; Josse, R.G.; Gomis, R.; Hanefeld, M.; Karasik, A.; Laakso, M.; Group, S.N.T.R. Acarbose treatment and the risk of cardiovascular disease and hypertension in patients with impaired glucose tolerance. J. Am. Med. Assoc. 2003, 290, 486–494. [Google Scholar] [CrossRef] [PubMed]
  5. Kawamori, R.; Tajima, N.; Iwamoto, Y.; Kashiwagi, A.; Shimamoto, K.; Kaku, K.; Group, V.P.S. Voglibose for prevention of type 2 diabetes mellitus: A randomised, double-blind trial in Japanese individuals with impaired glucose tolerance. Lancet 2009, 373, 1607–1614. [Google Scholar] [CrossRef]
  6. Monge, A.; Palop, J.; Ramierz, C.; Font, M.; Fernandez-Alveraz, E. New 5H-1,2,4-triazino[5,6-b]indole and aminoindole derivatives. Synthesis and studies as inhibitors of blood platelet aggregation, anti-hypertensive agents and thromboxane synthetase inhibitorsNouveaux dérivés 5H-1,2,4-triazino[5,6-b]indole et de l’aminoindole. Synthèse et évaluation comme inhibiteurs de l’agrégation plaquettaire, agents anti-hypertenseurs et inhibiteurs de la thromboxane synthétase. Eur. J. Med. Chem. 1991, 26, 179. [Google Scholar]
  7. Shelke, S.M.; Bhosale, S.H. Synthesis, antidepressant evaluation and QSAR studies of novel 2-(5H-[1,2,4]triazino[5,6-b]indol-3-ylthio)-N-(substituted phenyl)acetamides. Bioorg. Med. Chem. Lett. 2010, 20, 4661–4664. [Google Scholar] [CrossRef]
  8. Tomchin, A.B.; Uryupov, O.Y.; Zhukova, T.I.; Kuznetsova, T.A.; Kostycheva, M.V.; Smirnov, A.V. Thiourea and Thiosemicarbazide Derivatives: Structure, Transformations, and Pharmacological Activity. Part II. Antihypoxic Activity of 1,2,4-triazino[5,6-b]indole Derivatives. Pharmaceut. Chem. J. 1997, 31, 125–133. [Google Scholar] [CrossRef]
  9. Aswar, U.M.; Kalshetti, P.P.; Shelke, S.M.; Bhosale, S.H.; Bodhankar, S.L. Effect of newly synthesized 1,2,4-triazino[5,6-b]indole-3-thione derivatives on olfactory bulbectomy induced depression in rats. Asian Pac. J. Trop. Biomed. 2012, 2, 992. [Google Scholar] [CrossRef]
  10. Kgokong, J.L.; Smith, P.P.; Matsabisa, G.M. 1,2,4-Triazino-[5,6b]indole derivatives: Effects of the trifluoromethyl group on in vitro antimalarial activity. Bioorg. Med. Chem. 2005, 13, 2935–2942. [Google Scholar] [CrossRef]
  11. Gladych, J.M.Z.; Hunt, J.H.; Jack, D.; Haff, R.F.; Boyle, J.J.; Stewart, R.C.; Ferlanto, R.J. Inhibition of Rhinovirus by Isatin Thiosemicarbazone Analogues. Nature 1969, 221, 286. [Google Scholar] [CrossRef] [PubMed]
  12. Boyle, J.J.; Raupp, W.G.; Stanfield, F.J.; Haff, R.F.; Dick, E.C.; Alessio, D.D.; Dick, C.R. Progress in Rhinovirus Chenotherapy. Ann. N. Y. Acad. Sci. 1970, 173, 477. [Google Scholar] [CrossRef]
  13. Gwaltney, J.M. Rhinovirus inhibition by 3-substituted triazinoindoles. Proc. Soc. Exp. Biol. Med. 1970, 133, 1148. [Google Scholar] [CrossRef] [PubMed]
  14. Haff, R.F.; Flagg, W.B.; Gallo, J.J.; Hoover, J.R.E.; Miller, J.A.; Pinto, C.A.; Pagano, J.F. The in vitro antiviral activity of a triazinoindole (SK & F 40491). Proc. Soc. Exp. Biol. Med. 1972, 141, 475–478. [Google Scholar]
  15. Noreen, T.; Taha, M.; Imran, S.; Chigurpati, S.; Rahim, F.; Selvaraj, M.; Ismail, N.H.; Mohammad, J.I.; Ullah, H.; Javid, M.T.; et al. Synthesis of Alpha Amylase Inhibitors Based on Privileged Indole Scaffold. Bioorg. Chem. 2017, 72, 248–255. [Google Scholar] [CrossRef]
  16. Rahim, F.; Ali, M.; Ullah, S.; Rashid, U.; Ullah, H.; Taha, M.; Javed, M.T.; Rehman, W.; Abid, O.U.R.; Khan, A.A.; et al. Development of bis-Thiobarbiturates as Successful Urease Inhibitors and their Molecular Modeling Studies. Chin. Chem. Lett. 2016, 27, 693–697. [Google Scholar] [CrossRef]
  17. Taha, M.; Sultan, S.; Nuzar, H.A.; Rahim, F.; Imran, S.; Ismail, N.H.; Naz, H.; Ullah, H. Synthesis and biological evaluation of novel N-arylidenequinoline-3-carbohydrazides as potent β-glucuronidase inhibitors. Bioorg. Med. Chem. 2016, 24, 3696–3704. [Google Scholar] [CrossRef]
  18. Zaman, K.; Rahim, F.; Taha, M.; Ullah, H.; Wadood, A.; Nawaz, M.; Khan, F.; Wahab, Z.; Shah, S.A.A.; Rehman, A.; et al. Synthesis, in vitro urease inhibitory potential and molecular docking study of Benzimidazole analogues. Bioorg. Chem. 2019, 89, 103024. [Google Scholar] [CrossRef]
  19. Rashid, U.; Rahim, F.; Taha, M.; Arshad, M.; Ullah, H.; Mahmood, T.; Ali, M. Synthesis of 2-Acylated and Sulfonated 4-hydroxycoumarins: In vitro Urease Inhibition and Molecular Docking Studies. Bioorg. Chem. 2016, 66, 111–116. [Google Scholar] [CrossRef]
  20. Khan, K.M.; Rasheed, H.; Fatima, B.; Hayat, M.; Rahim, F.; Ullah, H.; Hameed, A.; Taha, M.; Tahir, A.; Perveen, S. Anti-Cancer Potential of Benzophenone-Bis-Schiff bases on Human Pancreatic Cancer Cell Line. J. Chem. Soc. Pak. 2016, 38, 954–958. [Google Scholar]
  21. Taha, M.; Ismail, N.H.; Imran, S.; Rahim, F.; Wadood, A.; Khan, H.; Ullah, H.; Salar, U.; Khan, K.M. Synthesis, β-Glucuronidase Inhibition and Molecular Docking Studies of Hybrid Bisindole-Thiosemicarbazides Analogs. Bioorg. Chem. 2016, 68, 56–63. [Google Scholar] [CrossRef] [PubMed]
  22. Rahim, F.; Javid, M.T.; Ullah, H.; Wadood, A.; Taha, M.; Ashraf, M.; Aine, Q.U.; Khan, M.A.; Khan, F.; Mirza, S.; et al. Synthesis, Molecular Docking, Acetylcholinesterase and Butyrylcholinesterase Inhibitory Potential of Thiazole Analogs as New Inhibitors for Alzheimer Disease. Bioorg. Chem. 2015, 62, 106–116. [Google Scholar] [CrossRef] [PubMed]
  23. Ullah, H.; Rahim, F.; Taha, M.; Hussain, R.; Wadood, A.; Nawaz, M.; Wahab, Z.; Kanwal; Khan, K.M. Synthesis, In vitro α-Glucosidase Inhibitory Potential and Molecular Docking Studies of 2-Amino-1,3,4-Oxadiazole Derivatives. Med. Chem. 2019, 15. [Google Scholar] [CrossRef] [PubMed]
  24. Rahim, F.; Ullah, H.; Taha, M.; Wadood, A.; Javid, M.T.; Rehman, W.; Nawaz, M.; Ashraf, M.; Ali, M.; Sajid, M.; et al. Synthesis and in vitro Acetylcholinesterase and Butyrylcholinesterase Inhibitory Potential of Hydrazide based Schiff Bases. Bioorg. Chem. 2016, 68, 30–40. [Google Scholar] [CrossRef] [PubMed]
  25. Rahim, F.; Taha, M.; Ullah, H.; Wadood, A.; Selvaraj, M.; Rab, A.; Sajid, M.; Shah, S.A.A.; Uddin, N.; Gollapalli, M. Synthesis of new arylhydrazide bearing Schiff bases/thiazolidinone: α-amylase, urease activities and their molecular docking studies. Bioorg. Chem. 2019, 91, 103112. [Google Scholar] [CrossRef]
  26. Gollapalli, M.; Taha, M.; Ullah, H.; Nawaz, M.; Al Muqarrabun, L.M.R.; Rahim, F.; Qureshi, F.; Mosaddik, A.; Ahmat, N.; Khan, K.M. Synthesis of Bis-indolylmethane sulfonohydrazides derivatives as potent α-Glucosidase inhibitors. Bioorg. Chem. 2018, 80, 112–120. [Google Scholar] [CrossRef]
  27. Ullah, H.; Rahim, F.; Taha, M.; Uddin, I.; Wadood, A.; Shah, S.A.A.; Farooq, R.K.; Nawaz, M.; Wahab, Z.; Khan, K.M. Synthesis, Molecular docking study and in vitro Thymidine Phosphorylase Inhibitory Potential of Oxadiazole Derivatives. Bioorg. Chem. 2018, 78, 58–67. [Google Scholar] [CrossRef]
  28. Taha, M.; Ullah, H.; Al Muqarrabun, L.M.R.; Khan, M.N.; Rahim, F.; Ahmat, N.; Javid, M.T.; Ali, M.; Khan, K.M. Bisindolylmethane thiosemicarbazides as potential inhibitors of urease: Synthesis and molecular modeling studies. Bioorg. Med. Chem. 2018, 26, 152–160. [Google Scholar] [CrossRef]
  29. Taha, M.; Ullah, H.; Al Muqarrabun, L.M.R.; Khan, M.N.; Rahim, F.; Ahmat, N.; Ali, M.; Perveen, S. Synthesis of bis-indolylmethanes as new potential inhibitors of β-glucuronidase and their molecular docking studies. Eur. J. Med. Chem. 2018, 143, 1757–1767. [Google Scholar] [CrossRef]
  30. Rahim, F.; Ullah, K.; Ullah, H.; Wadood, A.; Taha, M.; Rehman, A.; din, I.U.; Ashraf, M.; Shaukat, A.; Rehman, W.; et al. Triazinoindole analogs as potent inhibitors of α-glucosidase: Synthesis, biological evaluation and molecular docking studies. Bioorg. Chem. 2015, 58, 81–87. [Google Scholar] [CrossRef]
  31. Imran, S.; Taha, M.; Ismail, N.H.; Kashif, S.M.; Jamil, W.; Hariono, M.; Yusuf, M.; Wahab, H.; Imran, S. Synthesis of novel flavone hydrazones: In-vitro evaluation of α-glucosidase inhibition, QSAR analysis and docking studies. Eur. J. Med. Chem. 2015, 105, 156–170. [Google Scholar] [CrossRef] [PubMed]
  32. Rahim, F.; Ullah, H.; Javid, M.T.; Wadood, A.; Taha, M.; Ashraf, M.; Shaukat, A.; Junaid, M.; Hussain, S.; Rehman, W.; et al. Synthesis, in vitro evaluation and molecular docking studies of thiazole derivatives as new inhibitors of α-glucosidase. Bioorg. Chem. 2015, 62, 15–21. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are available from the authors.
Figure 1. Rationale of the current work.
Figure 1. Rationale of the current work.
Molecules 24 03819 g001
Scheme 1. Synthesis of triazinoindole derivatives.
Scheme 1. Synthesis of triazinoindole derivatives.
Molecules 24 03819 sch001
Scheme 2. Synthesis of thiosemicarbazide derivatives.
Scheme 2. Synthesis of thiosemicarbazide derivatives.
Molecules 24 03819 sch002
Scheme 3. Synthesis of triazinoindole-based thiosemicarbazide analogs (125).
Scheme 3. Synthesis of triazinoindole-based thiosemicarbazide analogs (125).
Molecules 24 03819 sch003
Figure 2. Validation through superimposition of (a) acarbose in 3W37 and (b) α-d-glucopyranose in 3A4A.
Figure 2. Validation through superimposition of (a) acarbose in 3W37 and (b) α-d-glucopyranose in 3A4A.
Molecules 24 03819 g002
Figure 3. Docking position of compound 1 in the active site of the α-glucosidase enzyme.
Figure 3. Docking position of compound 1 in the active site of the α-glucosidase enzyme.
Molecules 24 03819 g003
Figure 4. Docking position of compound 23 in the active site of the α-glucosidase enzyme.
Figure 4. Docking position of compound 23 in the active site of the α-glucosidase enzyme.
Molecules 24 03819 g004
Figure 5. Docking position of compound 16 in the active site of the α-glucosidase enzyme.
Figure 5. Docking position of compound 16 in the active site of the α-glucosidase enzyme.
Molecules 24 03819 g005
Table 1. Structure of triazinoindole-based thiosemicarbazide analogs and their α-glucosidase activity.
Table 1. Structure of triazinoindole-based thiosemicarbazide analogs and their α-glucosidase activity.
S. No.RR1IC50 (μM)
1 Molecules 24 03819 i001 Molecules 24 03819 i0021.30 ± 0.05
2 Molecules 24 03819 i001 Molecules 24 03819 i0032.60 ± 0.05
3 Molecules 24 03819 i004 Molecules 24 03819 i0027.5 ± 0.20
4 Molecules 24 03819 i004 Molecules 24 03819 i00612.80 ± 0.30
5 Molecules 24 03819 i001 Molecules 24 03819 i0065.20 ± 0.30
6 Molecules 24 03819 i004 Molecules 24 03819 i00725.3 ± 0.50
7 Molecules 24 03819 i006 Molecules 24 03819 i00733.30 ± 0.60
8 Molecules 24 03819 i006 Molecules 24 03819 i00617.5 ± 0.30
9 Molecules 24 03819 i008 Molecules 24 03819 i0096.80 ± 0.10
10 Molecules 24 03819 i001 Molecules 24 03819 i00735.80 ± 0.80
11 Molecules 24 03819 i010 Molecules 24 03819 i00726.80 ± 0.70
12 Molecules 24 03819 i010 Molecules 24 03819 i0068.30 ± 0.20
13 Molecules 24 03819 i004 Molecules 24 03819 i01123.30 ± 0.50
14 Molecules 24 03819 i010 Molecules 24 03819 i0117.5 ± 0.20
15 Molecules 24 03819 i001 Molecules 24 03819 i0115.80 ± 0.20
16 Molecules 24 03819 i001 Molecules 24 03819 i0131.8 ± 0.20
17 Molecules 24 03819 i012 Molecules 24 03819 i0098.80 ± 0.20
18 Molecules 24 03819 i014 Molecules 24 03819 i0152.30 ± 0.05
19 Molecules 24 03819 i004 Molecules 24 03819 i0152.30 ± 0.05
20 Molecules 24 03819 i006 Molecules 24 03819 i0117.50 ± 0.20
21 Molecules 24 03819 i006 Molecules 24 03819 i0154.80 ± 0.20
22 Molecules 24 03819 i014 Molecules 24 03819 i0165.90 ± 0.10
23 Molecules 24 03819 i010 Molecules 24 03819 i0051.30 ± 0.01
24 Molecules 24 03819 i004 Molecules 24 03819 i00915.3 ± 0.30
25 Molecules 24 03819 i014 Molecules 24 03819 i0099.30 ± 0.30
Acarbose38.60 ± 0.20
Table 2. Chemscore value for active compounds 1, 16, and 23.
Table 2. Chemscore value for active compounds 1, 16, and 23.
CompoundsChemscore (kJ/mol)IC50 (μM ± SD)
1−89.31.30 ± 0.05
16−74.51.80 ± 0.20
23−87.71.30 ± 0.01

Share and Cite

MDPI and ACS Style

Taha, M.; Alshamrani, F.J.; Rahim, F.; Hayat, S.; Ullah, H.; Zaman, K.; Imran, S.; Khan, K.M.; Naz, F. Synthesis of Novel Triazinoindole-Based Thiourea Hybrid: A Study on α-Glucosidase Inhibitors and Their Molecular Docking. Molecules 2019, 24, 3819. https://doi.org/10.3390/molecules24213819

AMA Style

Taha M, Alshamrani FJ, Rahim F, Hayat S, Ullah H, Zaman K, Imran S, Khan KM, Naz F. Synthesis of Novel Triazinoindole-Based Thiourea Hybrid: A Study on α-Glucosidase Inhibitors and Their Molecular Docking. Molecules. 2019; 24(21):3819. https://doi.org/10.3390/molecules24213819

Chicago/Turabian Style

Taha, Muhammad, Foziah J. Alshamrani, Fazal Rahim, Shawkat Hayat, Hayat Ullah, Khalid Zaman, Syahrul Imran, Khalid Mohammed Khan, and Farzana Naz. 2019. "Synthesis of Novel Triazinoindole-Based Thiourea Hybrid: A Study on α-Glucosidase Inhibitors and Their Molecular Docking" Molecules 24, no. 21: 3819. https://doi.org/10.3390/molecules24213819

Article Metrics

Back to TopTop