1
|
Peytam F, Hosseini FS, Fathimolladehi R, Nayeri MJD, Moghadam MS, Bayati B, Norouzbahari M, Foroumadi R, Bonyasi F, Divsalar R, Mojtabavi S, Faramarzi MA, Tehrani MB, Firoozpour L, Foroumadi A. Design, synthesis, and evaluation of novel substituted imidazo[1,2-c]quinazoline derivatives as potential α-glucosidase inhibitors with bioactivity and molecular docking insights. Sci Rep 2024; 14:27507. [PMID: 39528585 PMCID: PMC11555253 DOI: 10.1038/s41598-024-78878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
α-Glucosidase inhibitors are important in the treatment of type 2 diabetes by regulating blood glucose levels and reducing carbohydrate absorption. The present study focuses on identifying new inhibitors bearing imidazo[1,2-c]quinazoline backbone through multi-step synthesis. The inhibitory potencies of the novel derivatives were tested against Saccharomyces cerevisiae α-glucosidase, revealing IC50 values ranging from 50.0 ± 0.12 µM to 268.25 ± 0.09 µM. Among them, 2-(4-(((2,3-diphenylimidazo[1,2-c]quinazolin-5-yl)thio)methyl)-1H-1,2,3-triazol-1-yl)-N-(2-methoxyphenyl)acetamide (19e) and 2-(4-((benzo[4,5]imidazo[1,2-c]quinazolin-6-ylthio)methyl)-1H-1,2,3-triazol-1-yl)-N-(2-methoxyphenyl)acetamide (27e) emerged as the most potent inhibitors and were further investigated in various assessments. Finally, molecular docking studies were performed to reveal the crucial binding interactions and to confirm the results obtained from structure-activity relationship (SAR) analysis.
Collapse
Affiliation(s)
- Fariba Peytam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Sadat Hosseini
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Fathimolladehi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahdis Sadeghi Moghadam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Bayati
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Norouzbahari
- Faculty of Pharmacy, Final International University, Catalkoy, Kyrenia via Mersin 10 Turkey, Turkish Republic of Northern Cyprus
| | - Roham Foroumadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Bonyasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ruzbehan Divsalar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Barazandeh Tehrani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2
|
Bodala V, Podugu RL, Yettula K, Kadiri SK, Vidavalur S. Substituent-Oriented Synthesis of Substituted Pyridines/Pyrido[3,2-C]coumarins via Sequential Reactions of α-H and Me/Aryl Substituted Oxime Acetates and 3-Formylchromones. J Org Chem 2024; 89:12957-12966. [PMID: 39196990 DOI: 10.1021/acs.joc.4c00899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
A facile one pot sequential and highly chemoselective synthesis of substituted pyridines/pyrido[3,2-c]coumarins has been developed from oxime acetates and 3-formylchromones in the presence of FeCl2. This reaction was oriented by different substituents on the α-position (H, methyl/aryl) of oxime acetates. Mechanistic investigations suggested that substituted pyridines were formed via intramolecular aza-Michael addition followed by ring opening, while pyrido[3,2-c]coumarins were formed via intermolecular Michael addition. Besides, anti-TB activity was screened for some of the synthesized pyridines. Among the tested compounds, 3ga, 3ha, 3ja, 3ma, 3ac, 4pa, 4pb and 4sb were found to have good activity with an MIC of 12.5 μg/mL.
Collapse
Affiliation(s)
- Varaprasad Bodala
- Department of Organic Chemistry and FDW, School of Chemistry, AU College of Science and Technology, Andhra University, Visakhapatnam, India, 500003
| | - Rajitha Lakshmi Podugu
- Department of Organic Chemistry and FDW, School of Chemistry, AU College of Science and Technology, Andhra University, Visakhapatnam, India, 500003
| | - Kumari Yettula
- Department of Organic Chemistry and FDW, School of Chemistry, AU College of Science and Technology, Andhra University, Visakhapatnam, India, 500003
| | - Sunanda Kumari Kadiri
- Department of MicrobiologyAndhra University, AU College of Science and Technology, Andhra University, Visakhapatnam, India, 530003
| | - Siddaiah Vidavalur
- Department of Organic Chemistry and FDW, School of Chemistry, AU College of Science and Technology, Andhra University, Visakhapatnam, India, 500003
| |
Collapse
|
3
|
Sepehri S, Farhadi G, Maghbul M, Nasiri F, Faramarzi MA, Mahnam K, Mojtabavi S, Mahdavi M, Moharrami Oranj Z. A new route to the synthesis of 2-hydrazolyl-4-thiazolidinone hybrids, evaluation of α-glucosidase inhibitory activity and molecular modeling insights. Heliyon 2024; 10:e36408. [PMID: 39247345 PMCID: PMC11380029 DOI: 10.1016/j.heliyon.2024.e36408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
One of the multifactorial worldwide health syndromes is diabetes mellitus which is increasing at a disturbing rate. The inhibition of α-glucosidase, an enzyme that catalyzes starch hydrolysis in the intestine, is one helpful therapeutic approach for controlling hyperglycemia related to type-2 diabetes. To discover α-glucosidase inhibitors, some 2-hydrazolyl-4-thiazolidinone hybrids (3a-e) were synthesized from new one-pot reaction procedures. Next, their chemical structures were confirmed by 1H NMR, 13C NMR, and FT-IR spectra, and elemental analysis technique. Then, the α-glucosidase inhibitory activity of the titled compounds was evaluated. Among them, derivatives 3b and 3c revealed the highest activity against α-glucosidase compared to acarbose as a drug. Enzyme kinetic studies of the most active derivative (3b) indicated a competitive inhibition. Finally, molecular modeling studies were accomplished to describe vital interactions of the most potent compounds (3b and 3c) with the α-glucosidase enzyme.
Collapse
Affiliation(s)
- Saghi Sepehri
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghazaleh Farhadi
- Students Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Maghbul
- Department of Applied Chemistry, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Farough Nasiri
- Department of Applied Chemistry, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Karim Mahnam
- Department of Biology, Faculty of Sciences, Shahrekord University, Shahrekord, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
4
|
Gopi B, Vijayakumar V. An efficient and simple approach for synthesizing indazole compounds using palladium-catalyzed Suzuki-Miyaura cross-coupling. RSC Adv 2024; 14:26494-26504. [PMID: 39175677 PMCID: PMC11339776 DOI: 10.1039/d4ra04633a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
A series of indazole derivatives (6a-6i and 7a-7i) has been synthesized using Suzuki Miyaura cross-coupling with a palladium catalyst from readily available starting materials. An efficient and reliable methodology was employed for the synthesis, and the compounds were thoroughly characterized using 1H NMR, 13C NMR, FT-IR, and HRMS analysis to confirm their structural integrity and purity. Density function theory (DFT) computation identified four compounds (6g, 6h, 7g, and 7h) with significant energy band gaps. Additionally, the molecular electrostatic potential study highlighted the distinct electrical characteristics of these indazole molecules. Subsequent molecular docking investigations were carried out using the AUTODOCK method with two separate protein data bank (PDB) structures (6FEW, 4WA9) involved in renal cancer pathways. The results showed that eight substances PDB: 6FEW (6g, 6h, 7g, and 7h) and PDB: 4WA9 (6a, 6c, and 7c, 7g) had the highest binding energies, indicating their potential as therapeutic agents for treating kidney cancer.
Collapse
Affiliation(s)
- Bandaru Gopi
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology Vellore 632014 India
| | | |
Collapse
|
5
|
El-Sofany WI, Azzam EMS, Latif S, Hamden K. Spirothiazolidine-Derivative on Silver Nanoparticles and Carbon Nanotubes: Evaluation of Antibacterial, Anti-Fungal, Anti-Inflammatory, Antioxidant and Gastroprotective Activities. Pharmaceutics 2024; 16:901. [PMID: 39065599 PMCID: PMC11279549 DOI: 10.3390/pharmaceutics16070901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
This study aims to develop innovative heterocyclic nanocomposites incorporating silver nanoparticles (SNPs) for potential therapeutic applications targeting infections, gastric ulceration, inflammation, and oxidative damage. By synthesizing new derivatives of spiro-thiazolidine-carbonitrile (Py-ST-X) and incorporating them into Ag nanoparticles (AgNPs) and carbon nanotubes (CNTs), we have prepared Ag@Py-ST-X and Ag@Py-ST-X@CNT nanocomposites, respectively. The physical properties of these materials were studied using XRD, TEM, SEM, and Zeta potential techniques. In our investigation involving rats with gastric ulcers, we observed noteworthy inhibitory effects on gastric acid enzyme activity, specifically H+/K+ATPase, by Ag@Py-ST-NO2 and Ag@Py-ST-Br nanocomposites, demonstrating reductions of 25 and 34%, respectively, compared to untreated ulcers. Nanotubulation of these compounds further improved their inhibitory efficacy to 29 and 45%, respectively. Additionally, these nanoparticles showed the most potent myeloperoxidase (MPO)-inhibitory activity, demonstrating 36 and 49% inhibition, respectively, with nanotubulated versions reaching 44 and 53%. Moreover, Ag@Py-ST-NO2@CNT and Ag@Py-ST-Br@CNT nanotubes showed significant antioxidant activity, reducing thiobarbituric acid reactive substances (TBARS) by 35 and 51%, and hydrogen peroxide (H2O2) levels by 49 and 71%, respectively. These therapeutic effects were confirmed by reductions in gastric surface area (GSA) by 44% and 52%, a decrease in ulcer index (UI) from 80% to 44 and 38%, and an increase in curative index (CI) from 19 to 55 and 62% following administration of Ag@Py-ST-NO2@CNT and Ag@Py-ST-Br@CNT, respectively. Histological studies support these findings, suggesting the potential of these nanocomposites as promising candidates for treating various disorders.
Collapse
Affiliation(s)
- Walaa I. El-Sofany
- Department of Chemistry, College of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
- Photochemistry Department, Chemical Industries Research Institute, National Research Centre, 33 EL Buhouth St., Dokki, Giza 12622, Egypt
| | - Eid. M. S. Azzam
- Department of Chemistry, College of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
- Egyptian Petroleum Research Institute, Nasr City, Cairo 11727, Egypt
| | - Salman Latif
- Department of Chemistry, College of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
| | - Khaled Hamden
- Laboratory of Bioresources: Integrative Biology and Exploiting, Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir 5000, Tunisia
| |
Collapse
|
6
|
Alshammari OAO, Azzam EMS, Alhar MS, Alanazi KD, Aljuhani SAA, Elsofany WI. Antibacterial and Anticandidal Activity of the Nanostructural Composite of a Spirothiazolidine-Derivative Assembled on Silver Nanoparticles. Molecules 2024; 29:1139. [PMID: 38474650 DOI: 10.3390/molecules29051139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/17/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
Our aims in this work are the preparation of an ionic liquid based on heterocyclic compounds with Ag nanoparticles and the investigation of its application as an antibacterial and anticandidal agent. These goals were achieved through the fabrication of an ionic liquid based on Ag nanoparticles with 5-Amino-3-(4-fluorophenyl)-N-hexadecyl-7-(4-methylphenyl)-2-H spiro[cyclohexane1,2'-[1,3]thiazolo [4,5-b]pyridine]-6-carbonitrile (P16). The nanostructure of the prepared ionic liquid was characterized using techniques such as FTIR, 1HNMR, 13CNMR, UV, SEM, and TEM. The biological activity of the prepared compound (P16) and its nanocomposites with Ag nanoparticles was tested using five clinical bacteria (Pseudomonas aeruginosa 249; Escherichia coli 141; Enterobacter cloacae 235; Staphylococcus epidermidis BC 161, and methicillin-resistant S. aureus 217), and three Candida species (Candida utilis ATCC 9255; C. tropicalis ATCC 1362, and C. albicans ATCC 20402). The FTIR, 1HNMR, and 13CNMR results confirmed the chemical structure of the synthesized P16 compound. The nanostructure of the prepared ionic liquid was determined based on data obtained from the UV, SEM, and TEM tests. The antibacterial and anticandidal results showed that the biological activity of the compound (P16) was enhanced after the formation of nanocomposite structures with Ag nanoparticles. Moreover, the biological activity of the compound itself (P16) and that of its nanocomposite structure with Ag nanoparticles was higher than that of ampicillin and amphotericin B, which were used as control drugs in this work.
Collapse
Affiliation(s)
- Odeh A O Alshammari
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia
| | - Eid M S Azzam
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia
- Egyptian Petroleum Research Institute, Nasr City, Cairo 11727, Egypt
| | - Munirah S Alhar
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia
| | - Kaseb D Alanazi
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia
| | - Sara A A Aljuhani
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia
| | - Walaa I Elsofany
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia
- Photochemistry Department, Chemical Industries Research Institute, National Research Centre, 33 EL Buhouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|
7
|
Ghomi MK, Dastyafteh N, Montazer MN, Noori M, Mojtabavi S, Faramarzi MA, Hashemi SM, Mahdavi M. Synthesis, in vitro potency of inhibition, enzyme kinetics and in silico studies of quinoline-based α-glucosidase inhibitors. Sci Rep 2024; 14:501. [PMID: 38177164 PMCID: PMC10766639 DOI: 10.1038/s41598-023-50711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/23/2023] [Indexed: 01/06/2024] Open
Abstract
Diabetes mellitus is a multifactorial global health disorder that is rising at an alarming rate. One effective therapeutic approach for controlling hyperglycemia associated with type-2 diabetes is to target α-glucosidase, which catalyzes starch hydrolysis in the intestine. In an attempt to find potential α-glucosidase inhibitors, a series of twenty new quinoline linked benzothiazole hybrids (8a-t) were synthesized in good yields from suitable reaction procedures and their chemical structures were analyzed by 1HNMR, 13CNMR, IR, and ESI-MS analysis. The synthesized derivatives further screened for their activity against α-glucosidase. Among them, compounds 8b, 8h, 8n and 8o exhibited remarkable α-glucosidase inhibitory activity with IC50 values ranging from 38.2 ± 0.3 to 79.9 ± 1.2 µM compared with standard drug acarbose (IC50 = 750.0 ± 2.0 µM). Enzyme kinetic studies of the most active compound (8h) indicated a non-competitive inhibition with Ki value of 38.2 µM. Moreover, the homology modeling, molecular docking and molecular dynamics simulation studies were conducted to reveal key interactions between the most active compound 8h and the targeted enzyme. These results are complementary to the experimental observations. In order to predict the druggability of the novel derivatives, the pharmacokinetic properties were also applied. These findings could be useful for the design and development of new α-glucosidase inhibitors.
Collapse
Affiliation(s)
- Minoo Khalili Ghomi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Navid Dastyafteh
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Nazari Montazer
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Noori
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Mahdieh Hashemi
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Nidhar M, Kumar V, Mahapatra A, Gupta P, Yadav BK, Singh RK, Tewari AK. Ligand-based designing of DPP-4 inhibitors via hybridization; synthesis, docking, and biological evaluation of pyridazine-acetohydrazides. Mol Divers 2023; 27:2729-2740. [PMID: 36534357 DOI: 10.1007/s11030-022-10577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022]
Abstract
A series of novel pyridazine-acetohydrazide hybrids were designed, synthesized, and evaluated for their in vitro and in vivo antihyperglycemic activity. In this context, pyridazine-acetohydrazides (6a-6p) were synthesized by coupling substituted aldehyde with 2-(5-cyano-6-oxo-3,4-diphenylpyridazine-1-6H-yl) acetohydrazide, which was prepared via the reaction of pyridazine ester with hydrazine hydrate. The molecular docking study was carried out to examine the binding affinities and interaction of designed compounds against the DPP-4 enzyme. Compounds 6e, 6f, 6l, and 6n exhibited interaction with active residue. In silico ADMET properties, and toxicity studies corroborated that compounds were found to have good bioavailability and less toxic. The synthesized compounds were further estimated for in vitro DPP-4 activity. Compounds 6e and 6l were found as the most effective DPP-4 inhibitor in this series with IC50 values (6.48, 8.22 nM) when compared with sitagliptin (13.02 nM). According to the toxicity assay compound, 6l showed very less toxicity at a higher concentration so further selected for the in vivo antihyperglycemic activity.
Collapse
Affiliation(s)
- Manisha Nidhar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Vipin Kumar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Archisman Mahapatra
- Molecular Endocrinology and Toxicology Lab (MET Lab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Priya Gupta
- Molecular Endocrinology and Toxicology Lab (MET Lab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Brijesh Kumar Yadav
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Rahul Kumar Singh
- Molecular Endocrinology and Toxicology Lab (MET Lab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| | - Ashish Kumar Tewari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
9
|
Shamim S, Khan KM, Ali M, Mahdavi M, Salar U, Mohammadi-Khanaposhtani M, Faramarzi MA, Ullah N, Taha M. Diphenyl-substituted triazine derivatives: synthesis, α-glucosidase inhibitory activity, kinetics and in silico studies. Future Med Chem 2023; 15:1651-1668. [PMID: 37727987 DOI: 10.4155/fmc-2023-0057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
Background: Diabetes mellitus (DM) is a chronic disorder, considered to be a major global health challenge in the 21st century. α-Glucosidase enzyme is a well-known drug target to treat Type II DM. Methods: A new library of biphenyl-substituted triazines was synthesized and confirmed by various spectroscopic techniques. Results: All compounds showed potent α-glucosidase inhibitory activity, with IC50 values ranging from 35.35 ± 0.34 to 564.41 ± 0.91 μM, as the standard acarbose, IC50 value of 750.7 ± 0.13 μM. Our in silico study has predicted key interactions with the enzyme's active site. Drug-likeness and absorption, distribution, metabolism, excretion and toxicity were also studied. Conclusion: This study has identified a range of potential hits against the α-glucosidase enzyme that may serve as antidiabetic agents after further investigations.
Collapse
Affiliation(s)
- Shahbaz Shamim
- HEJ Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Khalid Mohammed Khan
- HEJ Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Department of Clinical Pharmacy, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, PO Box 31441, Dammam, Saudi Arabia
| | - Muhammad Ali
- HEJ Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Mohammad Mahdavi
- Endocrinology & Metabolism Research Center, Endocrinology & Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Uzma Salar
- Dr. Panjwani Center for Molecular Medicine & Drug Research, International Center for Chemical & Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Maryam Mohammadi-Khanaposhtani
- Cellular & Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nisar Ullah
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, PO Box 31441, Dammam, Saudi Arabia
| |
Collapse
|
10
|
Kosuge S, Araki Y, Tsuge K, Sugimoto K, Matsuya Y. One-Pot Synthesis of Pentasubstituted Pyridines following the Gold(I)-Catalyzed Aza-Enyne Metathesis/6π-Electrocyclization-Aromatization Sequence. J Org Chem 2023. [PMID: 37191633 DOI: 10.1021/acs.joc.3c00270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The one-pot de novo synthesis of pentasubstituted pyridines was realized following the process of Au(I)-autotandem catalysis and subsequent aromatization. The process involves aza-enyne metathesis with aryl propiolates to yield 1-azabutadienes and their addition/6π-electrocyclization sequence with the other propiolate units. The resultant 1,4-dihydropyridines were aromatized to furnish the pyridines in the presence of atmospheric oxygen. The aryl propiolates were regioselectively incorporated into the ring system to afford 2-arylpyridines as the sole product.
Collapse
Affiliation(s)
- Shuto Kosuge
- Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Yusuke Araki
- Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kiyoshi Tsuge
- Faculty of Science, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Kenji Sugimoto
- Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Yuji Matsuya
- Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
11
|
Banda BP, Medishetti N, Nanubolu JB, Atmakur K. One-pot domino synthesis of five- and six-membered fused dihydropyridines promoted by PPh 3-NBS in aqueous medium. Org Biomol Chem 2023; 21:4434-4439. [PMID: 37183547 DOI: 10.1039/d3ob00472d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
A facile one-pot synthesis of five- and six-membered fused dihydropyridines such as chromenodihydropyridines, pyrazolodihydropyridines and isoxazolopyridines was accomplished for the first time by employing PPh3-NBS via a formal [3 + 2 + 1] cycloaddition of 1,3-bisnucleophiles (i.e., 2-aminochromone, 4-aminochromone, 5-aminopyrazole and 5-aminoisoxazole), β-enaminones and aldehydes in aqueous medium. The present approach involves a Michael type addition followed by intramolecular cyclization leading to the formation of two new C-C bonds and one C-N bond. High compatibility and excellent yields are the advantages of this protocol.
Collapse
Affiliation(s)
- Bhanu Prasad Banda
- Fluoro & Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, India.
| | - Nagaraju Medishetti
- Fluoro & Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Jagadeesh Babu Nanubolu
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
- Laboratory of X-ray Crystallography, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500 007, India
| | - Krishnaiah Atmakur
- Fluoro & Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
12
|
Saleem F, Haider M, Khan KM, Özil M, Baltaş N, Ul-Haq Z, Qureshi U, Salar U, Taha M, Hameed S, Ullah N. Regioselective syntheses of 2-oxopyridine carbonitrile derivatives and evaluation for antihyperglycemic and antioxidant potential. Int J Biol Macromol 2023; 241:124589. [PMID: 37116840 DOI: 10.1016/j.ijbiomac.2023.124589] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
A library of 2-oxopyridine carbonitriles 1-34 was synthesized by regioselective nucleophilic substitution reactions. In the first step, a one-pot multicomponent reaction yield pyridone intermediates. The resulting pyridone intermediates were then reacted with phenacyl halides in DMF and stirred at 100 °C for an hour to afford the desired compounds in good yields. Structures of synthetic molecules were characterized by EI-MS, HREI-MS, 1H NMR, and 13C NMR, and all thirty-four (34) compounds were found to be new. All synthetic compounds were examined for antidiabetic and antioxidant potential. The compounds exhibited α-glucosidase inhibitory potential in the range of IC50 = 3.00 ± 0.11-43.35 ± 0.67 μM and α-amylase inhibition potential in the range of IC50 = 9.20 ± 0.14-65.56 ± 1.05 μM. Among the tested compounds, 1 showed the most significant α-glucosidase inhibitory activity, with an IC50 value of 3.00 ± 0.11 μM, while the most active compound against α-amylase was 6, with an IC50 value = 9.20 ± 0.14 μM. The kinetic studies and analysis indicated that the compounds followed the competitive mode of inhibition. In addition, the molecular docking studies showed the interaction profile of all molecules with the binding site residues of α-glucosidase and α-amylase enzymes.
Collapse
Affiliation(s)
- Faiza Saleem
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Maham Haider
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 31441, Dammam, Saudi Arabia.
| | - Musa Özil
- Department of Chemistry, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Nimet Baltaş
- Department of Chemistry, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Urooj Qureshi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Uzma Salar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 31441, Dammam, Saudi Arabia
| | - Shehryar Hameed
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Nisar Ullah
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| |
Collapse
|
13
|
Ramkumar S, Ramarajan R. Design, Synthesis, Spectral Characterization, Antioxidant Activity, Molecular Docking and in silico ADMET Studies of 1, 3 Oxazepines. ChemistrySelect 2023. [DOI: 10.1002/slct.202204818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Affiliation(s)
- Subramaniyan Ramkumar
- Department of Chemistry Annamalai University, Annamalai Nagar Tamil Nadu 608 002 India
| | - Rajalakshmi Ramarajan
- Department of Chemistry Annamalai University, Annamalai Nagar Tamil Nadu 608 002 India
| |
Collapse
|
14
|
Odeh AA, Al-Jaber HI, Barhoumi LM, Al-Fawares O, Shakya AK, Al-Qudah MA, Sanabra O. Phytochemical and bioactivity evaluation of secondary metabolites and essential oils of Sedum rubens growing wild in Jordan. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
15
|
|
16
|
Design, synthesis, spectral characterization, multiple-biological activities, docking and in silico ADMET studies of thiazolidinones. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2023.100861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
17
|
Novel Set of Highly Substituted Bis-pyridines: Synthesis, Molecular Docking and Drug-Resistant Antibacterial Profile. Future Med Chem 2022; 14:1881-1897. [PMID: 36420816 DOI: 10.4155/fmc-2022-0196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aims: Development of antimicrobial agents having the ability to prevent bacterial biofilm formation which causes serious health problems, especially with antibiotic-resistant bacterial strains. Materials and methods: The use of 1,3-diaryl enones as structural motifs to access the pyridine core. Antimicrobial activities of the synthesized compounds against methicillin-susceptible Staphylococcus aureus, methicillin-resistant S. aureus and vancomycin-resistant S. aureus bacterial strains were investigated. Results: The newly synthesized bis-enones were used as building blocks to access some novel highly substituted bis-pyridine derivatives. Several novel bis-compounds showed great bacterial biofilm eradication activity. Conclusion: A new series of bis-chalcones was synthesized and their structural diversity was exploited to access the corresponding, more biologically active, pyridine core. These bis-pyridines showed respectable antibacterial activities against various drug-resistant bacterial strains: namely, methicillin-susceptible, methicillin-resistant and vancomycin-resistant S. aureus.
Collapse
|
18
|
Hu CM, Luo YX, Wang WJ, Li JP, Li MY, Zhang YF, Xiao D, Lu L, Xiong Z, Feng N, Li C. Synthesis and Evaluation of Coumarin-Chalcone Derivatives as α-Glucosidase Inhibitors. Front Chem 2022; 10:926543. [PMID: 35832461 PMCID: PMC9271751 DOI: 10.3389/fchem.2022.926543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Coumarin and chalcone, two important kinds of natural product skeletons, both exhibit α-glucosidase inhibitory activity. In this work, coumarin-chalcone derivatives 3 (a∼v) were synthesized, and their α-glucosidase inhibitory activity was screened. The results showed that all synthetic derivatives (IC50: 24.09 ± 2.36 to 125.26 ± 1.18 μM) presented better α-glucosidase inhibitory activity than the parent compounds 3-acetylcoumarin (IC50: 1.5 × 105 μM) and the positive control acarbose (IC50: 259.90 ± 1.06 μM). Among them, compound 3t displayed the highest α-glucosidase inhibitory activity (IC50: 24.09 ± 2.36 μM), which was approximately 10 times stronger than that of acarbose. The kinetic assay of 3t (KI = 18.82 μM, KIS = 59.99 μM) revealed that these compounds inhibited α-glucosidase in a mixed-type manner. Molecular docking was used to simulate the interaction between α-glucosidase and compound 3t.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhuang Xiong
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| | - Na Feng
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| | - Chen Li
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| |
Collapse
|
19
|
Chen Z, Zeng P, Zhang S, Sun J. Recent Advances in Organic Synthesis of 3-Amino- or 4-Aminocoumarins. MINI-REV ORG CHEM 2022. [DOI: 10.2174/1570193x18666211001124004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Coumarin is a privileged scaffold that contains the unique 2H-chromen-2-one motif, and its
derivatives are widely distributed in nature, especially in plants. In recent years, due to their diverse
pharmacological activities and remarkable photochemical properties, they have attracted significant
attention from scientists, which has also prompted the research on the synthesis approaches and the
availability of substrates for these compounds. This article is a brief description of the methods for the
synthesis of various coumarin derivatives via two- or multi-component reactions involving 3-amino
or 4-aminocoumarin reported during 2015-2021. This review may help expand the development of
various analogues with coumarin as the basic unit.
Collapse
Affiliation(s)
- Zhiwei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Piaopiao Zeng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Shuo Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Jie Sun
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| |
Collapse
|
20
|
Fallah Z, Tajbakhsh M, Alikhani M, Larijani B, Faramarzi MA, Hamedifar H, Mohammadi-Khanaposhtani M, Mahdavi M. A review on synthesis, mechanism of action, and structure-activity relationships of 1,2,3-triazole-based α-glucosidase inhibitors as promising anti-diabetic agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132469] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
21
|
De S, Kumar S K A, Shah SK, Kazi S, Sarkar N, Banerjee S, Dey S. Pyridine: the scaffolds with significant clinical diversity. RSC Adv 2022; 12:15385-15406. [PMID: 35693235 PMCID: PMC9121228 DOI: 10.1039/d2ra01571d] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/27/2022] [Indexed: 12/20/2022] Open
Abstract
The nitrogen-bearing heterocycle pyridine in its several analogous forms occupies an important position as a precious source of clinically useful agents in the field of medicinal chemistry research. This privileged scaffold has been consistently incorporated in a diverse range of drug candidates approved by the FDA (Food and Drug Administration). This moiety has attracted increasing attention from several disease states owing to its ease of parallelization and testing potential pertaining to the chemical space. In the next few years, a larger share of novel pyridine-based drug candidates is expected. This review unifies the current advances in novel pyridine-based molecular frameworks and their unique clinical relevance as reported over the last two decades. It highlights an inclination to the use of pyridine-based molecules in drug crafting and the subsequent emergence of several potent and eligible candidates against a range of diversified diseases. The nitrogen-bearing heterocycle pyridine in its several analogous forms occupies an important position as a precious source of clinically useful agents in the field of medicinal chemistry research.![]()
Collapse
Affiliation(s)
- Sourav De
- Department of Pharmaceutical Technology, School of Medical Science, Adamas University, Kolkata-700126, West Bengal, India
| | - Ashok Kumar S K
- Department of Chemistry, School of Advanced Sciences, VIT, Vellore-632014, India
| | - Suraj Kumar Shah
- Department of Pharmaceutical Technology, School of Medical Science, Adamas University, Kolkata-700126, West Bengal, India
| | - Sabnaz Kazi
- Department of Pharmaceutical Technology, School of Medical Science, Adamas University, Kolkata-700126, West Bengal, India
| | - Nandan Sarkar
- Department of Pharmaceutical Technology, School of Medical Science, Adamas University, Kolkata-700126, West Bengal, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol-713301, West Bengal, India
| | - Sanjay Dey
- Department of Pharmaceutical Technology, School of Medical Science, Adamas University, Kolkata-700126, West Bengal, India
| |
Collapse
|
22
|
Design, synthesis of coumarin tethered 1,2,3-triazoles analogues, evaluation of their antimicrobial and α-amylase inhibition activities. J CHEM SCI 2021. [DOI: 10.1007/s12039-021-01997-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Tashrifi Z, Mohammadi-Khanaposhtani M, Larijani B, Hamedifar H, Ansari S, Mahdavi M. Vinylazides: versatile synthons and magical precursors for the construction of N-heterocycles. Mol Divers 2021; 25:2533-2570. [PMID: 32474890 DOI: 10.1007/s11030-020-10106-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/15/2020] [Indexed: 10/24/2022]
Abstract
Nowadays, application of vinylazides as precursors is a key method for the construction of N-heterocycles in organic synthesis. These versatile three-atom synthons can be converted into intermediates such as 2H-azirines, iminyl radicals, iminyl metal complexes, iminyl inions and nitrilium ions that subsequently afford a wide range of polyfunctional cyclic nitrogen-containing compounds. In this review, the reactions of vinylazides leading to these products (in the last decade) are categorized based on the types of the resulting N-heterocyclic rings and a brief and concise description of the reaction mechanisms is presented.
Collapse
Affiliation(s)
- Zahra Tashrifi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Mohammadi-Khanaposhtani
- College of Engineering, Faculty of Fouman, University of Tehran, Fouman, Iran
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Halleh Hamedifar
- College of Engineering, Faculty of Fouman, University of Tehran, Fouman, Iran
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Samira Ansari
- College of Engineering, Faculty of Fouman, University of Tehran, Fouman, Iran.
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
24
|
Efficient synthesis of novel chromenopyrido[3,2-e]isothiazolo[2,3-a]pyrimidines via a non-catalytic one-pot three-component reaction. RESEARCH ON CHEMICAL INTERMEDIATES 2021. [DOI: 10.1007/s11164-021-04613-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
25
|
Ling Y, Hao ZY, Liang D, Zhang CL, Liu YF, Wang Y. The Expanding Role of Pyridine and Dihydropyridine Scaffolds in Drug Design. Drug Des Devel Ther 2021; 15:4289-4338. [PMID: 34675489 PMCID: PMC8520849 DOI: 10.2147/dddt.s329547] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022] Open
Abstract
Pyridine-based ring systems are one of the most extensively used heterocycles in the field of drug design, primarily due to their profound effect on pharmacological activity, which has led to the discovery of numerous broad-spectrum therapeutic agents. In the US FDA database, there are 95 approved pharmaceuticals that stem from pyridine or dihydropyridine, including isoniazid and ethionamide (tuberculosis), delavirdine (HIV/AIDS), abiraterone acetate (prostate cancer), tacrine (Alzheimer's), ciclopirox (ringworm and athlete's foot), crizotinib (cancer), nifedipine (Raynaud's syndrome and premature birth), piroxicam (NSAID for arthritis), nilvadipine (hypertension), roflumilast (COPD), pyridostigmine (myasthenia gravis), and many more. Their remarkable therapeutic applications have encouraged researchers to prepare a larger number of biologically active compounds decorated with pyridine or dihydropyridine, expandeing the scope of finding a cure for other ailments. It is thus anticipated that myriad new pharmaceuticals containing the two heterocycles will be available in the forthcoming decade. This review examines the prospects of highly potent bioactive molecules to emphasize the advantages of using pyridine and dihydropyridine in drug design. We cover the most recent developments from 2010 to date, highlighting the ever-expanding role of both scaffolds in the field of medicinal chemistry and drug development.
Collapse
Affiliation(s)
- Yong Ling
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Zhi-You Hao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, People’s Republic of China
| | - Dong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, People’s Republic of China
| | - Chun-Lei Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yan-Fei Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yan Wang
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Gummidi L, Kerru N, Ebenezer O, Awolade P, Sanni O, Islam MS, Singh P. Multicomponent reaction for the synthesis of new 1,3,4-thiadiazole-thiazolidine-4-one molecular hybrids as promising antidiabetic agents through α-glucosidase and α-amylase inhibition. Bioorg Chem 2021; 115:105210. [PMID: 34332231 DOI: 10.1016/j.bioorg.2021.105210] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/29/2021] [Accepted: 07/22/2021] [Indexed: 01/07/2023]
Abstract
A simple and efficient protocol was developed to synthesize a new library of thiazolidine-4-one molecular hybrids (4a-n) via a one-pot multicomponent reaction involving 5-substituted phenyl-1,3,4-thiadiazol-2-amines, substituted benzaldehydes and 2-mercaptoacetic acid. The synthesized compounds were evaluated in vitro for their antidiabetic activities through α-glucosidase and α-amylase inhibition as well as their antioxidant and antimicrobial potentials. Compound 4e exhibited the most promising α-glucosidase and α-amylase inhibition with an IC50 value of 2.59 μM, which is ~1.5- and 14-fold superior as compared to the standard inhibitor acarbose. Structure-activity relationship (SAR) analysis revealed that the nature and position of substituents on the phenyl rings had a significant effect on the inhibitory potency.
Collapse
Affiliation(s)
- Lalitha Gummidi
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Nagaraju Kerru
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Oluwakemi Ebenezer
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Paul Awolade
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Olakunle Sanni
- Department of Biochemistry, School of Life Sciences, University of Kwazulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of Kwazulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Parvesh Singh
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa.
| |
Collapse
|
27
|
Bushra, Shamim S, Khan KM, Ullah N, Mahdavi M, Faramarzi MA, Larijani B, Salar U, Rafique R, Taha M, Perveen S. Synthesis, in vitro, and in silico evaluation of Indazole Schiff bases as potential α-glucosidase inhibitors. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
4-(Trifluoromethyl) coumarin-fused pyridines: Regioselective synthesis and photophysics, electrochemical, and antioxidative activity. J Fluor Chem 2021. [DOI: 10.1016/j.jfluchem.2021.109822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Mollazadeh M, Mohammadi-Khanaposhtani M, Valizadeh Y, Zonouzi A, Faramarzi MA, Kiani M, Biglar M, Larijani B, Hamedifar H, Mahdavi M, Hajimiri MH. Novel Coumarin Containing Dithiocarbamate Derivatives as Potent α-Glucosidase Inhibitors for Management of Type 2 Diabetes. Med Chem 2021; 17:264-272. [PMID: 32851964 DOI: 10.2174/1573406416666200826101205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND α-Glucosidase is a hydrolyzing enzyme that plays a crucial role in the degradation of carbohydrates and starch to glucose. Hence, α-glucosidase is an important target in carbohydrate mediated diseases such as diabetes mellitus. OBJECTIVE In this study, novel coumarin containing dithiocarbamate derivatives 4a-n were synthesized and evaluated against α-glucosidase in vitro and in silico. METHODS These compounds were obtained from the reaction between 4-(bromomethyl)-7- methoxy-2H-chromen-2-one 1, carbon disulfide 2, and primary or secondary amines 3a-n in the presence of potassium hydroxide and ethanol at room temperature. In vitro α-glucosidase inhibition and kinetic study of these compounds were performed. Furthermore, a docking study of the most potent compounds was also performed by Auto Dock Tools (version 1.5.6). RESULTS Obtained results showed that all the synthesized compounds exhibited prominent inhibitory activities (IC50 = 85.0 ± 4.0-566.6 ± 8.6 μM) in comparison to acarbose as a standard inhibitor (IC50 = 750.0 ± 9.0 μM). Among them, the secondary amine derivative 4d with pendant indole group was the most potent inhibitor. Enzyme kinetic study of the compound 4d revealed that this compound competes with a substrate to connect to the active site of α-glucosidase and therefore is a competitive inhibitor. Moreover, a molecular docking study predicted that this compound interacted with the α-glucosidase active site pocket. CONCLUSION Our results suggest that the coumarin-dithiocarbamate scaffold can be a promising lead structure for designing potent α-glucosidase inhibitors for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Marjan Mollazadeh
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Yousef Valizadeh
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Afsaneh Zonouzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad A Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Kiani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Hamed Hajimiri
- Nano Alvand Company, Avicenna Tech Park, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Peytam F, Takalloobanafshi G, Saadattalab T, Norouzbahari M, Emamgholipour Z, Moghimi S, Firoozpour L, Bijanzadeh HR, Faramarzi MA, Mojtabavi S, Rashidi-Ranjbar P, Karima S, Pakraad R, Foroumadi A. Design, synthesis, molecular docking, and in vitro α-glucosidase inhibitory activities of novel 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines against yeast and rat α-glucosidase. Sci Rep 2021; 11:11911. [PMID: 34099819 PMCID: PMC8184976 DOI: 10.1038/s41598-021-91473-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
In an attempt to find novel, potent α-glucosidase inhibitors, a library of poly-substituted 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines 3a-ag have been synthesized through heating a mixture of 2-aminobenzimidazoles 1 and α-azidochalcone 2 under the mild conditions. This efficient, facile protocol has been resulted into the desirable compounds with a wide substrate scope in good to excellent yields. Afterwards, their inhibitory activities against yeast α-glucosidase enzyme were investigated. Showing IC50 values ranging from 16.4 ± 0.36 µM to 297.0 ± 1.2 µM confirmed their excellent potency to inhibit α-glucosidase which encouraged us to perform further studies on α-glucosidase enzymes obtained from rat as a mammal source. Among various synthesized 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines, compound 3k exhibited the highest potency against both Saccharomyces cerevisiae α-glucosidase (IC50 = 16.4 ± 0.36 μM) and rat small intestine α-glucosidase (IC50 = 45.0 ± 8.2 μM). Moreover, the role of amine moiety on the observed activity was studied through substituting with chlorine and hydrogen resulted into a considerable deterioration on the inhibitory activity. Kinetic study and molecular docking study have confirmed the in-vitro results.
Collapse
Affiliation(s)
- Fariba Peytam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Toktam Saadattalab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Norouzbahari
- Faculty of Medicine, Eastern Mediterranean University, via Mersin 10, Famagusta, Northern Cyprus, Turkey
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Bijanzadeh
- Department of Environmental Sciences, Faculty of Natural Resources and Marine Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Roya Pakraad
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Volochnyuk DM, Ryabukhin SV, Mityuk AP, Hrebonkin A, Lebed PS, Grabchuk GP. Efficient Route for the Synthesis of Diverse Heteroannelated 5-Cyanopyridines. SYNTHESIS-STUTTGART 2021. [DOI: 10.1055/a-1360-9852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AbstractThe new efficient, convenient protocol for the synthesis of heteroannelated 3-cyanopyridines and pyrimidines starting from diverse aminoheterocycles and 3,3-dimethoxy-2-formylpropionitrile sodium salt was elaborated. The advantages and improvements of the procedure compared to previously known methods are shown. The scope and limitations of the method are determined. The impact of the structural features on regioselectivity are discussed. The preparativeness, scalability, and application scope of the elaborated protocol are demonstrated by the synthesis of five heteroannelated 3-cyanopyridines in quantities up to 100 grams.
Collapse
Affiliation(s)
- Dmitriy M. Volochnyuk
- Enamine Ltd
- Taras Shevchenko National University of Kyiv
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine
| | | | | | | | | | | |
Collapse
|
32
|
New 4,5-diphenylimidazole-acetamide-1,2,3-triazole hybrids as potent α-glucosidase inhibitors: synthesis, in vitro and in silico enzymatic and toxicity evaluations. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02779-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Abstract
4-Aminocoumarins represent an important class of a versatile scaffold in organic synthesis and have been consistently used as a building block in organic chemistry for the synthesis of different heterocyclic compounds.
Collapse
Affiliation(s)
- Madieh Sadeghpour
- Department of Chemistry
- Takestan Branch
- Islamic Azad University
- Takestan
- Iran
| | - Abolfazl Olyaei
- Department of Chemistry
- Payame Noor University (PNU)
- Tehran
- Iran
| | - Alireza Adl
- Department of Chemistry
- Payame Noor University (PNU)
- Tehran
- Iran
| |
Collapse
|
34
|
Patra P, Kar GK. The synthesis, biological evaluation and fluorescence study of chromeno[4,3- b]pyridin/quinolin-one derivatives, the backbone of natural product polyneomarline C scaffolds: a brief review. NEW J CHEM 2021. [DOI: 10.1039/d0nj04761a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review presents the synthesis, biological and fluorescence study of chromeno[4,3-b]pyridin/quinolin-ones via classical reactions including metal-catalyzed and green reaction protocols.
Collapse
Affiliation(s)
- Prasanta Patra
- Department of Chemistry
- Jhargram Raj College
- Jhargram 721507
- India
| | | |
Collapse
|
35
|
Patra P. 4-Chloro-3-formylcoumarin as a multifaceted building block for the development of various bio-active substituted and fused coumarin heterocycles: a brief review. NEW J CHEM 2021. [DOI: 10.1039/d1nj02755g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This review presents the diverse synthesis of 3,4-substituted coumarins and 5-, 6- and 7-membered ring fused coumarins using 4-chloro-3-formylcoumarin as the precursor via classical reactions including metal-catalyzed and green reaction protocols.
Collapse
Affiliation(s)
- Prasanta Patra
- Department of Chemistry
- Jhargram Raj College
- Jhargram 721507
- India
| |
Collapse
|
36
|
Peytam F, Adib M, Shourgeshty R, Mohammadi-Khanaposhtani M, Jahani M, Imanparast S, Faramarzi MA, Moghadamnia AA, Larijani B, Mahdavi M. Synthesis and biological evaluation of new dihydroindolizino[8,7-b]indole derivatives as novel α-glucosidase inhibitors. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
37
|
Maurya AK, Mulpuru V, Mishra N. Discovery of Novel Coumarin Analogs against the α-Glucosidase Protein Target of Diabetes Mellitus: Pharmacophore-Based QSAR, Docking, and Molecular Dynamics Simulation Studies. ACS OMEGA 2020; 5:32234-32249. [PMID: 33376861 PMCID: PMC7758891 DOI: 10.1021/acsomega.0c03871] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/10/2020] [Indexed: 05/13/2023]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease, the third killer of mankind. The finding of potent drugs against diabetes remains challenging. In the present study, coumarin derivatives with known biological activity against diabetic protein have been used to predict functional groups' positions on coumarin derivatives. α-Glucosidase is a brush border membrane-bound lysosomal enzyme from the hydrolase enzyme family. It plays an important role in the metabolism of glycoproteins. Inhibitors of lysosomal α-glucosidase can reduce postprandial hyperglycemia. Due to this, lysosomal α-glucosidase is a good therapeutic target for drugs. A total of 116 coumarin derivatives with IC50 values against lysosomal α-glucosidase were selected for a CADD (computer-aided drug design) approach to identify more potent drugs. Pharmacophore modeling and atom-based 3-QSAR of 116 active compounds against lysosomal α-glucosidase were performed and identified positions and types of groups to increase activity. We performed molecular docking of 116 coumarin derivatives against the lysosomal α-glucosidase enzyme, and three compounds (isorutarine, 10_, and 36) resulted in a docking score of -7.64, -7.12, and -6.86 kcal/mol. The molecular dynamics simulation of the above three molecules and protein complex performed for 100 ns supported the interaction stability of isorutarine, 10_, and 36 with the lysosomal binding site α-glucosidase.
Collapse
|
38
|
Mollazadeh M, Mohammadi-Khanaposhtani M, Valizadeh Y, Zonouzi A, Faramarzi MA, Hariri P, Biglar M, Larijani B, Hamedifar H, Mahdavi M, Sepehri N. 2,4-Dioxochroman Moiety Linked to 1,2,3-triazole Derivatives as Novel α-glucosidase Inhibitors: Synthesis, In vitro Biological Evaluation, and Docking Study. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200802181634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, a novel series of 2,4-dioxochroman-1,2,3-triazole hybrids 8a-l
was synthesized by click reaction. These compounds were screened against α-glucosidase
through in vitro and in silico evaluations. All the synthesized hybrids exhibited excellent
α-glucosidase inhibition in comparison to standard drug acarbose. Representatively,
3-((((1-(3,4-dichlorobenzyl)-1H-1,2,3-triazol-4-yl)methyl)amino)methylene)chroman-2,4-
dione 8h with IC50 = 20.1 ± 1.5 μM against α-glucosidase, was 37-times more potent than
acarbose. Enzyme kinetic study revealed that compound 8h was a competitive inhibitor
against α-glucosidase. In silico docking study on chloro derivatives 8h, 8g, and 8i were
also performed in the active site of α -glucosidase. Evaluations on obtained interaction
modes and binding energies of these compounds confirmed the results obtained through in
vitro α-glucosidase inhibition.
Collapse
Affiliation(s)
- Marjan Mollazadeh
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Yousef Valizadeh
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Afsaneh Zonouzi
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parsa Hariri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Sepehri
- Nano Alvand Company, Avicenna Tech Park, Tehran University of Medical Sciences, Tehran, 1439955991, Iran
| |
Collapse
|
39
|
Tafesse TB, Bule MH, Khoobi M, Faramarzi MA, Abdollahi M, Amini M. Coumarin-based Scaffold as α-glucosidase Inhibitory Activity: Implication for the Development of Potent Antidiabetic Agents. Mini Rev Med Chem 2020; 20:134-151. [PMID: 31553294 DOI: 10.2174/1389557519666190925162536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/15/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Delaying the absorption of glucose through α-glucosidase enzyme inhibition is one of the therapeutic approaches in the management of Type 2 diabetes, which can reduce the incidence of postprandial hyperglycemia. The existence of chronic postprandial hyperglycemia impaired the endogenous antioxidant defense by inducing oxidative stress-induced pancreatic β-cell destruction through uncontrolled generation of free radicals such as ROS, which in turn, leads to various macrovascular and microvascular complications. The currently available α -glucosidase inhibitors, for instance, acarbose, have some side effects such as hypoglycemia at higher doses, liver problems, meteorism, diarrhea, and lactic acidosis. Therefore, there is an urgent need to discover and develop potential α-glucosidase inhibitors. OBJECTIVE Based on suchmotifs, researchers are intrigued to search for the best scaffold that displays various biological activities. Among them, coumarin scaffold has attracted great attention. The compound and its derivatives can be isolated from various natural products and/or synthesized for the development of novel α-glucosidase inhibitors. RESULTS This study focused on coumarin and its derivatives as well as on their application as potent antidiabetic agents and has also concentrated on the structure-activity relationship. CONCLUSION This review describes the applications of coumarin-containing derivatives as α - glucosidase inhibitors based on published reports which will be useful for innovative approaches in the search for novel coumarin-based antidiabetic drugs with less toxicity and more potency.
Collapse
Affiliation(s)
- Tadesse Bekele Tafesse
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences-International Campus (IC-TUMS), Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,School of Pharmacy, College of Health & Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mohammed Hussen Bule
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences-International Campus (IC-TUMS), Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacy, College of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia
| | - Mehdi Khoobi
- Department of Pharmaceutical Biomaterials, Medical Biomaterials Research Center and The Institute of Pharmaceutical Sciences (TIPS), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Ataee‐Kachouei T, Nasr‐Esfahani M, Mohammadpoor‐Baltork I, Mirkhani V, Moghadam M, Tangestaninejad S, Notash B. Ce(IV) immobilized on halloysite nanotube–functionalized dendrimer (Ce(IV)–G2): A novel and efficient dendritic catalyst for the synthesis of pyrido[3,2‐
c
]coumarin derivatives. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5948] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Tahereh Ataee‐Kachouei
- Department of Chemistry, Catalysis Division University of Isfahan Isfahan 81746‐73441 Iran
| | | | | | - Valiollah Mirkhani
- Department of Chemistry, Catalysis Division University of Isfahan Isfahan 81746‐73441 Iran
| | - Majid Moghadam
- Department of Chemistry, Catalysis Division University of Isfahan Isfahan 81746‐73441 Iran
| | | | - Behrouz Notash
- Department of Inorganic Chemistry and Catalysis Shahid Beheshti University General Campus, Evin Tehran 1983963113 Iran
| |
Collapse
|
41
|
Asgari MS, Mohammadi-Khanaposhtani M, Sharafi Z, Faramarzi MA, Rastegar H, Nasli Esfahani E, Bandarian F, Ranjbar Rashidi P, Rahimi R, Biglar M, Mahdavi M, Larijani B. Design and synthesis of 4,5-diphenyl-imidazol-1,2,3-triazole hybrids as new anti-diabetic agents: in vitro α-glucosidase inhibition, kinetic and docking studies. Mol Divers 2020; 25:877-888. [PMID: 32189236 DOI: 10.1007/s11030-020-10072-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 03/09/2020] [Indexed: 10/24/2022]
Abstract
Fourteen novel 4,5-diphenyl-imidazol-1,2,3-triazole hybrids 8a-n were synthesized with good yields by performing click reaction between the 4,5-diphenyl-2-(prop-2-yn-1-ylthio)-1H-imidazole and various benzyl azides. The synthesized compounds 8a-n were evaluated against yeast α-glucosidase, and all these compounds exhibited excellent inhibitory activity (IC50 values in the range of 85.6 ± 0.4-231.4 ± 1.0 μM), even much more potent than standard drug acarbose (IC50 = 750.0 μM). Among them, 4,5-diphenyl-imidazol-1,2,3-triazoles possessing 2-chloro and 2-bromo-benzyl moieties (compounds 8g and 8i) demonstrated the most potent inhibitory activities toward α-glucosidase. The kinetic study of the compound 8g revealed that this compound inhibited α-glucosidase in a competitive mode. Furthermore, docking calculations of these compounds were performed to predict the interaction mode of the synthesized compounds in the active site of α-glucosidase. A novel series of 4,5-diphenyl-imidazol-1,2,3-triazole hybrids 8a-n was synthesized with good yields by performing click reaction between the 4,5-diphenyl-2-(prop-2-yn-1-ylthio)-1Himidazole and various benzyl azides. The synthesized compounds 8a-n were evaluated against yeast α-glucosidase and all these compounds exhibited excellent inhibitory activity (IC50 values in the range of 85.6 ± 0.4-231.4 ± 1.0 μM), even much more potent than standard drug acarbose (IC50 = 750.0 μM).
Collapse
Affiliation(s)
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Zeinab Sharafi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Rastegar
- Food and Drug Research Institute, Food and Drug Administration, MOHE, Tehran, Iran
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bandarian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, University of Medical Sciences, Tehran, Iran
| | | | - Rahmatollah Rahimi
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Peytam F, Adib M, Shourgeshty R, Firoozpour L, Rahmanian-Jazi M, Jahani M, Moghimi S, Divsalar K, Faramarzi MA, Mojtabavi S, Safari F, Mahdavi M, Foroumadi A. An efficient and targeted synthetic approach towards new highly substituted 6-amino-pyrazolo[1,5-a]pyrimidines with α-glucosidase inhibitory activity. Sci Rep 2020; 10:2595. [PMID: 32054916 PMCID: PMC7018746 DOI: 10.1038/s41598-020-59079-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/16/2019] [Indexed: 01/10/2023] Open
Abstract
In an attempt to find novel α-glucosidase inhibitors, an efficient, straightforward reaction to synthesize a library of fully substituted 6-amino-pyrazolo[1,5-a]pyrimidines 3 has been investigated. Heating a mixture of α-azidochalcones 1 and 3-aminopyrazoles 2 under the mild condition afforded desired compounds with a large substrate scope in good to excellent yields. All obtained products were evaluated as α-glucosidase inhibitors and exhibited excellent potency with IC50 values ranging from 15.2 ± 0.4 µM to 201.3 ± 4.2 µM. Among them, compound 3d was around 50-fold more potent than acarbose (IC50 = 750.0 ± 1.5 µM) as standard inhibitor. Regarding product structures, kinetic study and molecular docking were carried out for two of the most potent ones.
Collapse
Affiliation(s)
- Fariba Peytam
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Adib
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran.
| | - Reihaneh Shourgeshty
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Rahmanian-Jazi
- Department of Medicinal Chemistry, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Jahani
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Setareh Moghimi
- Department of Medicinal Chemistry, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Kouros Divsalar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran. .,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
43
|
Yan DW, Huang CD, Zheng HH, Zhao N, Feng XL, Ma SJ, Zhang AL, Zhang Q. Meroterpene-Like α-Glucosidase Inhibitors Based on Biomimetic Reactions Starting from β-Caryophyllene. Molecules 2020; 25:molecules25020260. [PMID: 31936396 PMCID: PMC7024386 DOI: 10.3390/molecules25020260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Natural meroterpenes derived from phloroglucinols and β-caryophyllene have shown high inhibitory activity against α-glucosidase or cancer cells, however, the chemical diversity of this type of skeletons in Nature is limited. METHODS To expand the chemical space and explore their inhibitory activities against α-glucosidase (EC 3.2.1.20), we employed β-caryophyllene and some natural moieties (4-hydroxycoumarins, lawsone or syncarpic acid) to synthesize new types of meroterpene-like skeletons. All the products (including side products) were isolated and characterized by NMR, HR-MS, and ECD. RESULTS In total, 17 products (representing seven scaffolds) were generated through a one-pot procedure. Most products (12 compounds) showed more potential activity (IC50 < 25 μM) than the positive controls (acarbose and genistein, IC50 58.19, and 54.74 μM, respectively). Compound 7 exhibited the most potent inhibition of α-glucosidase (IC50 3.56 μM) in a mixed-type manner. The CD analysis indicated that compound 7 could bind to α-glucosidase and influence the enzyme's secondary structure. CONCLUSIONS Compound 7 could serve as a new type of template compound to develop α-glucosidase inhibitors. Full investigation of a biomimic reaction can be used as a concise strategy to explore diverse natural-like skeletons and search for novel lead compounds.
Collapse
Affiliation(s)
- Da-Wei Yan
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
| | - Cheng-Di Huang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
| | - Hang-Hang Zheng
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
| | - Na Zhao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
| | - Xiao-Lan Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China;
| | - Shuang-Jiang Ma
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
| | - An-Ling Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
- Correspondence: (A.-L.Z.); (Q.Z.); Tel.: +86-1809-272-0670 (Q.Z.)
| | - Qiang Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; (D.-W.Y.); (C.-D.H.); (H.-H.Z.); (N.Z.); (S.-J.M.)
- Correspondence: (A.-L.Z.); (Q.Z.); Tel.: +86-1809-272-0670 (Q.Z.)
| |
Collapse
|
44
|
Mohammadi‐Khanaposhtani M, Yahyavi H, Imanparast S, Harandi FN, Faramarzi MA, Foroumadi A, Larijani B, Biglar M, Mahdavi M. Benzoylquinazolinone derivatives as new potential antidiabetic agents: α‐Glucosidase inhibition, kinetic, and docking studies. J CHIN CHEM SOC-TAIP 2019. [DOI: 10.1002/jccs.201900268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Maryam Mohammadi‐Khanaposhtani
- Cellular and Molecular Biology Research CenterHealth Research Institute, Babol University of Medical Sciences Babol Iran
| | - Hoda Yahyavi
- Department of Medicinal ChemistryFaculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Somaye Imanparast
- Department of Pharmaceutical BiotechnologyFaculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences Tehran Iran
| | | | - Mohammad Ali Faramarzi
- Department of Pharmaceutical BiotechnologyFaculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Alireza Foroumadi
- Department of Medicinal ChemistryFaculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research CenterEndocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| | - Mahmood Biglar
- Endocrinology and Metabolism Research CenterEndocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research CenterEndocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
45
|
A one-pot and three-component synthetic approach for the preparation of asymmetric and multi-substituted 1,4-dihydropyrazines. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
46
|
Bakherad Z, Mohammadi-Khanaposhtani M, Sadeghi-Aliabadi H, Rezaei S, Fassihi A, Bakherad M, Rastegar H, Biglar M, Saghaie L, Larijani B, Mahdavi M. New thiosemicarbazide-1,2,3-triazole hybrids as potent α-glucosidase inhibitors: Design, synthesis, and biological evaluation. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.04.082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
47
|
Nasli‐Esfahani E, Mohammadi‐Khanaposhtani M, Rezaei S, Sarrafi Y, Sharafi Z, Samadi N, Faramarzi MA, Bandarian F, Hamedifar H, Larijani B, Hajimiri M, Mahdavi M. A new series of Schiff base derivatives bearing 1,2,3‐triazole: Design, synthesis, molecular docking, and α‐glucosidase inhibition. Arch Pharm (Weinheim) 2019; 352:e1900034. [DOI: 10.1002/ardp.201900034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/18/2019] [Accepted: 05/01/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Ensieh Nasli‐Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical Sciences Tehran Iran
| | - Maryam Mohammadi‐Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research InstituteBabol University of Medical Sciences Babol Iran
| | - Sepideh Rezaei
- School of PharmacyTabriz University of Medical Sciences Tabriz Iran
| | | | - Zeinab Sharafi
- Razi Herbal Medicines Research CenterLorestan University of Medical Sciences Khorramabad Iran
| | - Nasser Samadi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research CenterTehran University of Medical Sciences Tehran Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research CenterTehran University of Medical Sciences Tehran Iran
| | - Fatemeh Bandarian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical Sciences Tehran Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research CenterAlborz University of Medical Sciences Karaj Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical Sciences Tehran Iran
| | - Mirhamed Hajimiri
- Nano Alvand Company, Avicenna Tech ParkTehran University of Medical Sciences Tehran Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical Sciences Tehran Iran
| |
Collapse
|