1
|
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
|
2
|
Thabet HK, Abusaif MS, Imran M, Helal MH, Alaqel SI, Alshehri A, Mohd AA, Ammar YA, Ragab A. Discovery of novel 6-(piperidin-1-ylsulfonyl)-2H-chromenes targeting α-glucosidase, α-amylase, and PPAR-γ: Design, synthesis, virtual screening, and anti-diabetic activity for type 2 diabetes mellitus. Comput Biol Chem 2024; 111:108097. [PMID: 38772048 DOI: 10.1016/j.compbiolchem.2024.108097] [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: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024]
Abstract
A new series of 2H-chromene-based sulfonamide derivatives 3-12 has been synthesized and characterized using different spectroscopic techniques. The synthesized 2H-chromenes were synthesized by reacting activated methylene with 5-(piperidin-1-ylsulfonyl)salicylaldehyde through one-step condensation followed by intramolecular cyclization. Virtual screening of the designed molecules on α-glucosidase enzymes (PDB: 3W37 and 3A4A) exhibited good binding affinity suggesting that these derivatives may be potential α-glucosidase inhibitors. In-vitro α-glucosidase activity was conducted firstly at 100 µg/mL, and the results demonstrated good inhibitory potency with values ranging from 90.6% to 96.3% compared to IP = 95.8% for Acarbose. Furthermore, the IC50 values were determined, and the designed derivatives exhibited inhibitory potency less than 11 µg/mL. Surprisingly, two chromene derivatives 6 and 10 showed the highest potency with IC50 values of 0.975 ± 0.04 and 0.584 ± 0.02 µg/mL, respectively, compared to Acarbose (IC50 = 0.805 ± 0.03 µg/mL). Moreover, our work was extended to evaluate the in-vitro α-amylase and PPAR-γ activity as additional targets for diabetic activity. The results exhibited moderate activity on α-amylase and potency as PPAR-γ agonist making it a multiplet antidiabetic target. The most active 2H-chromenes 6 and 10 exhibited significant activity to PPAR-γ with IC50 values of 3.453 ± 0.14 and 4.653 ± 0.04 µg/mL compared to Pioglitazone (IC50 = 4.884±0.29 µg/mL) indicating that these derivatives improve insulin sensitivity by stimulating the production of small insulin-sensitive adipocytes. In-silico ADME profile analysis indicated compliance with Lipinski's and Veber's rules with excellent oral bioavailability properties. Finally, the docking simulation was conducted to explain the expected binding mode and binding affinity.
Collapse
Affiliation(s)
- Hamdy Khamees Thabet
- Department of Chemistry, College of Sciences and Arts, Northern Border University, Rafha 91911, Saudi Arabia.
| | - Moustafa S Abusaif
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohamed Hamdy Helal
- Department of Chemistry, College of Sciences and Arts, Northern Border University, Rafha 91911, Saudi Arabia
| | - Saleh Ibrahim Alaqel
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Ahmed Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, King Faisal Road, Dammam 31441, Saudi Arabia
| | - Abida Ash Mohd
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Yousry A Ammar
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | - Ahmed Ragab
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt.
| |
Collapse
|
3
|
Khan IU, Jamil Y, Khan A, Ahmad J, Iqbal A, Ali S, Hamayun M, Hussain A, Alrefaei AF, Almutairi MH, Ahmad A. Pichia pastoris Mediated Digestion of Water-Soluble Polysaccharides from Cress Seed Mucilage Produces Potent Antidiabetic Oligosaccharides. Pharmaceuticals (Basel) 2024; 17:704. [PMID: 38931372 PMCID: PMC11206588 DOI: 10.3390/ph17060704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Diabetes mellitus is a heterogeneous metabolic disorder that poses significant health and economic challenges across the globe. Polysaccharides, found abundantly in edible plants, hold promise for managing diabetes by reducing blood glucose levels (BGL) and insulin resistance. However, most of these polysaccharides cannot be digested or absorbed directly by the human body. Here we report the production of antidiabetic oligosaccharides from cress seed mucilage polysaccharides using yeast fermentation. The water-soluble polysaccharides extracted from cress seed mucilage were precipitated using 75% ethanol and fermented with Pichia pastoris for different time intervals. The digested saccharides were fractionated through gel permeation chromatography using a Bio Gel P-10 column. Structural analysis of the oligosaccharide fractions revealed the presence of galacturonic acid, rhamnose, glucuronic acid, glucose and arabinose. Oligosaccharide fractions exhibited the potential to inhibit α-amylase and α-glucosidase enzymes in a dose-dependent manner in vitro. The fraction DF73 exhibited strong inhibitory activity against α-amylase with IC50 values of 38.2 ± 1.12 µg/mL, compared to the positive control, acarbose, having an IC50 value of 29.18 ± 1.76 µg/mL. Similarly, DF72 and DF73 showed the highest inhibition of α-glucosidase, with IC50 values of 9.26 ± 2.68 and 50.47 ± 5.18 µg/mL, respectively. In in vivo assays in streptozotocin (STZ)-induced diabetic mice, these oligosaccharides significantly reduced BGL and improved lipid profiles compared to the reference drug metformin. Histopathological observations of mouse livers indicated the cytoprotective effects of these sugars. Taken together, our results suggest that oligosaccharides produced through microbial digestion of polysaccharides extracted from cress seed mucilage have the potential to reduce blood glucose levels, possibly through inhibition of carbohydrate-digesting enzymes and regulation of the various signaling pathways.
Collapse
Affiliation(s)
- Imdad Ullah Khan
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (I.U.K.); (Y.J.); (A.K.); (J.A.)
| | - Yusra Jamil
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (I.U.K.); (Y.J.); (A.K.); (J.A.)
| | - Aiman Khan
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (I.U.K.); (Y.J.); (A.K.); (J.A.)
| | - Jalwa Ahmad
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (I.U.K.); (Y.J.); (A.K.); (J.A.)
| | - Amjad Iqbal
- Department of Food Science and Technology, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Sajid Ali
- Department of Horticulture and Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Muhammad Hamayun
- Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (M.H.); (A.H.)
| | - Anwar Hussain
- Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (M.H.); (A.H.)
| | - Abdulwahed Fahad Alrefaei
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.F.A.); (M.H.A.)
| | - Mikhlid H. Almutairi
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.F.A.); (M.H.A.)
| | - Ayaz Ahmad
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (I.U.K.); (Y.J.); (A.K.); (J.A.)
| |
Collapse
|
4
|
Han L, Zhao D, Li Y, Jin J, El-Kott AF, Al-Saeed FA, Eldib AM. Assessment of the Anti-Breast Cancer Effects of Urolithin with Molecular Docking Studies in the In Vitro Condition: Introducing a Novel Chemotherapeutic Drug. Mol Biotechnol 2024; 66:554-566. [PMID: 37280483 DOI: 10.1007/s12033-023-00766-3] [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: 01/14/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023]
Abstract
A lot of research has been done on using natural items as diabetes treatment. The molecular docking study was conducted to evaluate the inhibitory activities of urolithin A against α-amylase, α-glucosidase, and aldose reductase. The molecular docking calculations indicated the probable interactions and the characteristics of these contacts at an atomic level. The results of the docking calculations showed the docking score of urolithin A against α-amylase was -5.169 kcal/mol. This value for α-glucosidase and aldose reductase was -3.657 kcal/mol and -7.635 kcal/mol, respectively. In general, the outcomes of the docking calculations revealed that urolithin A can construct several hydrogen bonds and hydrophobic contacts with the assessed enzymes and reduces their activities considerably. The properties of urolithin against common human breast cancer cell lines, i.e., SkBr3, MDA-MB-231, MCF-7, Hs578T, Evsa-T, BT-549, AU565 and 600MPE were evaluated. The IC50 of the urolithin was 400, 443, 392, 418, 397, 530, 566 and 551 against SkBr3, MDA-MB-231, MCF-7, Hs578T, Evsa-T, BT-549, AU565 and 600MPE, respectively. After doing the clinical trial studies, the recent molecule may be used as an anti-breast cancer supplement in humans. IC50 values of urolithin A on α-amylase, α-glucosidase, and aldose reductase enzymes were obtained at 16.14, 1.06 and 98.73 µM, respectively.
Collapse
Affiliation(s)
- Lu Han
- Department of General Surgery, Sijing Hospital of Songjiang District Shanghai, Shanghai, 201601, China
| | - Danbo Zhao
- Department of Oncology, Ezhou Central Hospital, Ezhou, 436000, Hubei, China
| | - Ya Li
- Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi, 710061, China
| | - Jianwei Jin
- Department of Oncology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Fatimah A Al-Saeed
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
| | - Ali M Eldib
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
- Alrayan Medical Colleges (AMC), Hejrah Street, P. O. Box 41411, Madinah, Kingdom of Saudi Arabia
| |
Collapse
|
5
|
Singh G, Singh R, Monga V, Mehan S. 3,5-Disubstituted-thiazolidine-2,4-dione hybrids as antidiabetic agents: Design, synthesis, in-vitro and In vivo evaluation. Eur J Med Chem 2024; 266:116139. [PMID: 38252989 DOI: 10.1016/j.ejmech.2024.116139] [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: 11/25/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
Diabetes is one of the fastest-growing metabolic disorders, nearly doubling the number of patients each year. There are different treatment approaches available for the management of diabetes, which lacks due to their side effects. The inhibition of enzymes involved in the metabolism of complex polysaccharides to monosaccharides has proven beneficial in patients with type 2 diabetes mellitus. Two enzymes, α-amylase and α-glucosidase, have emerged as potential drug targets and are widely explored for drug development against type 2 diabetes mellitus. In this context, thiazolidine-2,4-diones (TZDs) have emerged as potential drug candidates for developing newer molecules against α-amylase and α-glucosidase. Nineteen TZD-hybrids were synthesized and evaluated in vitro α-amylase and α-glucosidase inhibitory activity. The compounds 7i, 7k, and 7p have emerged as the best dual inhibitors with IC50 of 10.33 ± 0.11-20.94 ± 0.76 μM and 10.19 ± 0.25-24.07 ± 1.56 μM against α-glucosidase and α-amylase, respectively. The derivatives had good anti-oxidant activity, displaying IC50 = 14.95 ± 0.65-23.27 ± 0.99 μM. The compounds 7k and 7p showed the best inhibition of reactive oxygen species in the PNAC-1 cells. The molecules exhibit good binding within the active site of α-amylase (PDB id: 1B2Y) and α-glucosidase (PDB id: 3W37), displaying binding energies of -7.5 to -10.7 kcal/mol and -7.4 to -10.3 kcal/mol, respectively. Further, the compounds were nontoxic (LD50 = 500-1311 mg/kg) and possessed good GI absorption. The compounds 7i, 7k, and 7p were evaluated in vivo antidiabetic activity in an STZ-induced diabetic model in Wistar rats. The compound 7p emerged as the best compound in the in vivo studies; however, the activity was lesser than that of the standard drug pioglitazone.
Collapse
Affiliation(s)
- Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, Punjab, India; Research Scholar, IK Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Rajveer Singh
- Department of Pharmacognosy, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, Punjab, India
| | - Vikramdeep Monga
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, Punjab, India.
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, Punjab, India, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, India).
| |
Collapse
|
6
|
Khator R, Monga V. Recent advances in the synthesis and medicinal perspective of pyrazole-based α-amylase inhibitors as antidiabetic agents. Future Med Chem 2024. [PMID: 38230638 DOI: 10.4155/fmc-2023-0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024] Open
Abstract
Diabetes is a serious health threat across the globe, claiming millions of lives worldwide. Among the various strategies employed, inhibition of α-amylase is a therapeutic protocol for the management of Type 2 diabetes mellitus. α-Amylase is a crucial enzyme involved in the breakdown of dietary starch into simpler units. However, the clinically used α-amylase inhibitors have various drawbacks. Therefore, design and development of novel α-amylase inhibitors have gained significant attention. The pyrazole motif has been identified as a versatile scaffold in medicinal chemistry, and recent studies have led to the identification of various pyrazole-based α-amylase inhibitors. This review compiles therapeutic implications of pyrazole-appended α-amylase inhibitors; their synthesis, biological activities, structure-activity relationships and molecular docking studies are discussed.
Collapse
Affiliation(s)
- Rakesh Khator
- Drug Design & Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences & Natural Products, Central University of Punjab, VPO-Ghudda, 151401, Bathinda, Punjab, India
| | - Vikramdeep Monga
- Drug Design & Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences & Natural Products, Central University of Punjab, VPO-Ghudda, 151401, Bathinda, Punjab, India
| |
Collapse
|
7
|
Mitra S, Chatterjee S, Bose S, Panda P, Basak S, Ghosh N, Mandal SC, Singhmura S, Halder AK. Finding structural requirements of structurally diverse α-glucosidase and α-amylase inhibitors through validated and predictive 2D-QSAR and 3D-QSAR analyses. J Mol Graph Model 2024; 126:108640. [PMID: 37801809 DOI: 10.1016/j.jmgm.2023.108640] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/09/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by hyperglycemic state. The α-glucosidase and α-amylase are considered two major targets for the management of Type 2 DM due to their ability of metabolizing carbohydrates into simpler sugars. In the current study, cheminformatics analyses were performed to develop validated and predictive models with a dataset of 187 α-glucosidase and α-amylase dual inhibitors. Separate linear, interpretable and statistically robust 2D-QSAR models were constructed with datasets containing the activities of α-glucosidase and α-amylase inhibitors with an aim to explain the crucial structural and physicochemical attributes responsible for higher activity towards these targets. Consequently, some descriptors of the models pointed out the importance of specific structural moieties responsible for the higher activities for these targets and on the other hand, properties such as ionization potential and mass of the compounds as well as number of hydrogen bond donors in molecules were found to be crucial in determining the binding potentials of the dataset compounds. Statistically significant 3D-QSAR models were developed with both α-glucosidase and α-amylase inhibition datapoints to estimate the importance of 3D electrostatic and steric fields for improved potentials towards these two targets. Molecular docking performed with selected compounds with homology model of α-glucosidase and X-ray crystal structure of α-amylase largely supported the interpretations obtained from the cheminformatic analyses. The current investigation should serve as important guidelines for the design of future α-glucosidase and α-amylase inhibitors. Besides, the current investigation is entirely performed by using non-commercial open-access tools to ensure easy accessibility and reproducibility of the investigation which may help researchers throughout the world to work more on drug design and discovery.
Collapse
Affiliation(s)
- Soumya Mitra
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
| | - Subhadas Chatterjee
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
| | - Shobhan Bose
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
| | - Parthasarathi Panda
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
| | - Souvik Basak
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
| | - Nilanjan Ghosh
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Subhash C Mandal
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Saroj Singhmura
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
| | - Amit Kumar Halder
- Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India.
| |
Collapse
|
8
|
Soetan OA, Ajao FO, Ajayi AF. Blood glucose lowering and anti-oxidant potential of erythritol: An in vitro and in vivo study. J Diabetes Metab Disord 2023; 22:1217-1229. [PMID: 37975140 PMCID: PMC10638257 DOI: 10.1007/s40200-023-01237-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 05/09/2023] [Indexed: 11/19/2023]
Abstract
Background Diabetes is a major cause of death worldwide and Nigeria is not an exception. The quest to lower sugar levels has become a major factor in the management of diabetes; this has occasioned the use of substitutes for refined sugar in beverages. Erythritol is a natural sweetener gaining immense interest in recent times. Like metformin, erythritol has shown hydroxyl radical scavenging ability and has metabolic profile suitable for diabetics. Therefore, the blood glucose-lowering and anti-oxidant properties of erythritol under in vitro and in vivo systems were accessed. Methods Radical scavenging assay (ABTS and DPPH) and inhibition of carbohydrate digestive enzymes (alpha-amylase and alpha-glucosidase) were employed to determine in vitro anti-oxidant and glucose regulatory function of erythritol respectively. Molecular docking studies were performed between 3D structures of human pancreatic alpha-amylase and alpha-glucosidase, isomaltase from saccharomyces cerevisiae with erythritol. The drug-like activity of erythritol was also assessed.Thereafter, we investigated the effect of erythritol on blood glucose and antioxidant status of normal and streptozocin- nicotinamide-induced diabetes rats which were grouped into five (n = 5); Normal, Ery (normal and administered erythritol), Db (diabetic control), Db + Ery (diabetic and administered erythritol), and Db + Met (diabetic and administered metformin). Results Erythritol showed a considerable radical scavenging activity and an ability to inhibit alpha-amylase and alpha-glucosidase in vitro. Also, a significant reduction in glucose intolerance, blood glucose and hemoglobin A1c levels and improved antioxidant level was seen in erythritol-treated diabetic rats. Conclusion Erythritol showed anti-oxidant activity, alpha amylase and glucosidase enzyme inhibition property, improved antioxidant status and ameliorated blood glucose, HbA1c, and glucose intolerance following diabetes.
Collapse
Affiliation(s)
- O. A. Soetan
- Reproduction and Bionformatics Research Unit, Ladoke Akintola University of Technology, Ogbomosho, Oyo Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, Thomas Adewumi University, Oko, Kwara State Nigeria
| | - F. O. Ajao
- Reproduction and Bionformatics Research Unit, Ladoke Akintola University of Technology, Ogbomosho, Oyo Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo Nigeria
| | - A. F. Ajayi
- Reproduction and Bionformatics Research Unit, Ladoke Akintola University of Technology, Ogbomosho, Oyo Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo Nigeria
| |
Collapse
|
9
|
Viola, Muhammad N, Noor A, Sirajuddin M, Kubicki M, Rahim S, Samad A, Shujah S, Wadood A, Ali S. Designing and Exploration of the Biological Potentials of Novel Centrosymmetric Heteroleptic Copper(II) Carboxylates. Pharmaceuticals (Basel) 2023; 16:1462. [PMID: 37895933 PMCID: PMC10610105 DOI: 10.3390/ph16101462] [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: 08/20/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Copper(II) complexes with a general formula [Cu2(3,4-F2C6H3CH2COO)4(L)2], where L = 2-methylpyridine (1) and 3-methylpyridine (2), are reported here. The FTIR spectra of the complexes confirmed the bridging bidentate coordination mode of the carboxylate ligand. The low (475 and 449 cm-1) and strong (727 & 725 cm-1) intensity bands in the FTIR spectra, due to Cu-N stretches and pyridyl ring vibrations, confirmed coordination of the 2-/3-methyl pyridine co-ligands in complexes 1 and 2, respectively. A binuclear paddlewheel structural arrangement with a square pyramidal geometry was confirmed for copper atoms in the complexes via single-crystal X-ray analysis. The DPPH, •OH radical, and α-amylase enzyme inhibition assays showed higher activities for the complexes than for the free ligand acid. The binding constant (Kb = 1.32 × 105 for 1 and 5.33 × 105 for 2) calculated via UV-VIS absorption measurements and docking scores (-6.59 for 1 and -7.43 for 2) calculated via molecular docking showed higher SS-DNA binding potential for 2 compared to 1. Viscosity measurement also reflected higher DNA binding ability for 2 than 1. Both complexes 1 and 2 (docking scores of -7.43 and -6.95, respectively) were found to be more active inhibitors than the free ligand acid (docking score of -5.5159) against the target α-amylase protein. This in silico study has shown that the herein reported compounds follow the rules of drug-likeness and exhibit good potential for bioavailability.
Collapse
Affiliation(s)
- Viola
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (V.); (S.R.)
| | - Niaz Muhammad
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (V.); (S.R.)
| | - Awal Noor
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al-Hassa 31982, Saudi Arabia
| | - Muhammad Sirajuddin
- Department of Chemistry, University of Science and Technology Bannu, Bannu 28100, Pakistan;
| | - Maciej Kubicki
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznań, Poland;
| | - Shahnaz Rahim
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (V.); (S.R.)
| | - Abdus Samad
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (A.S.); (A.W.)
| | - Shaukat Shujah
- Department of Chemistry, Kohat University of Science & Technology, Kohat 26000, Pakistan;
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (A.S.); (A.W.)
| | - Saqib Ali
- Department of Chemistry, Quaid-I-Azam University Islamabad, Islamabad 45320, Pakistan;
| |
Collapse
|
10
|
Motavallihaghi S, Maghsood AH, Nematollahi D, Barati N, Asl SS, Farmani A, Foroughi-Parvar F, Fallah M. Dimedone nanoparticle as a promising approach against toxoplasmosis: In vitro and in vivo evaluation. Biomed Pharmacother 2023; 166:115356. [PMID: 37666178 DOI: 10.1016/j.biopha.2023.115356] [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: 07/14/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023] Open
Abstract
Toxoplasma gondii, an intracellular parasite, has shown drug resistance and therapeutic failure in recent years. Dimedone (DIM) has been introduced as a new chemical compound with anti-bacterial and anti-cancer properties. The aim of this study was to investigate the potential protective role of DIM nanoparticles in an animal model of toxoplasmosis. Cytotoxicity of DIM on Vero cell line assessed using MTT, and the effect of DIM on Toxoplasma gondii was evaluated by counting the number of parasites compared to the control group in vitro. The rate of pathogenesis and virulence of the parasite was checked on the liver cells of the animal model using hematoxylin-eosin staining. Furthermore, various parameters indicating oxidative stress were compared in mouse liver tissue in different groups. The release of the nanoparticle form was significantly longer than the free drugs. The IC50 of Nano-DIM was 60 µM and the reduction of intracellular parasite proliferation in the group Nano-DIM and Nano-PYR (Nano-primethamine) was significantly lower than the free drugs in vitro. Histopathology examination in the groups treated with dimedone nanomedicine showed that the degree of disintegration of the epithelium of the central vein of the liver and infiltration and vacuolization of liver cells were lower compared to the toxoplasmosis group. Additionally, the level of some oxidative stress indicators was observed to be lower in the nano-treated groups compared to other groups. The results of this study showed DIM can be used as a promising compound for anti-T. gondii activity and can prevent the proliferation of it in cells.
Collapse
Affiliation(s)
- Seyedmousa Motavallihaghi
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran
| | - Amir Hossein Maghsood
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran
| | - Davood Nematollahi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 65178-38683, the Islamic Republic of Iran
| | - Nastaran Barati
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran
| | - Sara Soleimani Asl
- Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran
| | - Abbas Farmani
- Dental Implant Research Center & Dental Research Center, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran
| | - Faeze Foroughi-Parvar
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran
| | - Mohammad Fallah
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, the Islamic Republic of Iran.
| |
Collapse
|
11
|
Synthesis and anti-α-glucosidase activity evaluation of betulinic acid derivatives. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
12
|
Recent developments in synthetic α-glucosidase inhibitors: A comprehensive review with structural and molecular insight. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
13
|
Fettach S, Thari FZ, Hafidi Z, Karrouchi K, Bouathmany K, Cherrah Y, El Achouri M, Benbacer L, El Mzibri M, Sefrioui H, Bougrin K, Faouzi MEA. Biological, toxicological and molecular docking evaluations of isoxazoline-thiazolidine-2,4-dione analogues as new class of anti-hyperglycemic agents. J Biomol Struct Dyn 2023; 41:1072-1084. [PMID: 34957934 DOI: 10.1080/07391102.2021.2017348] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In this work, three isoxazoline-thiazolidine-2,4-dione derivatives were synthesized and characterized by FT-IR, 1H-NMR, 13C-NMR and ESI-MS spectrometry. All compounds have been investigated for their α-amylase and α-glucosidase inhibitory activities. In vitro enzymatic evaluation revealed that all compounds were inhibitory potent against α-glucosidase with IC50 values varied from 40.67 ± 1.81 to 92.54 ± 0.43 µM, and α-amylase with IC50 in the range of 07.01 ± 0.02 to 75.10 ± 1.06 µM. One of the tested compounds were found to be more potent inhibitor compared to other compounds and standard drug Acarbose (IC50 glucosidase= 97.12 ± 0.35 µM and IC50 amylase= 2.97 ± 0.01 μM). All compounds were then evaluated for their acute toxicity in vivo and shown their safety at a high dose with LD > 2000mg/kg BW. A cell-based toxicity evaluation was performed to determine the safety of compounds on liver cells, using the MTT assay against HepG2 cells, and the results shown that all compounds have non-toxic impact against cell viability and proliferation compared to reference drug (Pioglitazone). Furthermore, the molecular homology analysis, SAR and the molecular binding properties of compound with the active site of α-amylase and α-glucosidase were confirmed through computational analysis. This study has identified the inhibitory potential of a new class of synthesized isoxazoline-thiazolidine-2,4-dione derivatives in controlling both hyperglycemia and type 2 diabetes mellitus without any hepatic toxicity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saad Fettach
- Laboratory of Pharmacology and Toxicology, Biopharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Fatima Zahra Thari
- Equipe de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Faculty of Science, B.P. 1014, Geophysics, Natural Patrimony and Green Chemistry (GEOPAC) Research Center, Mohammed V University in Rabat, Rabat, Morocco
| | - Zakaria Hafidi
- Department of Surfactants and Nanobiotechnology, IQAC-CSIC, c/Jordi Girona, Barcelona, Spain
| | - Khalid Karrouchi
- Laboratory of Analytical Chemistry and Bromatology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Kaoutar Bouathmany
- Biology and Molecular Research Unit, Department of Life Sciences, National Center for Energy, Nuclear Science and Technology (CNESTEN), Rabat, Morocco
| | - Yahia Cherrah
- Laboratory of Pharmacology and Toxicology, Biopharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Mohammed El Achouri
- Laboratoire de Physico-Chimie des Matériaux Inorganiques et Organiques, Centre des Sciences des Matériaux, Ecole Normale Supérieure-Rabat, Mohammed V University, Rabat, Morocco
| | - Laila Benbacer
- Biology and Molecular Research Unit, Department of Life Sciences, National Center for Energy, Nuclear Science and Technology (CNESTEN), Rabat, Morocco
| | - Mohammed El Mzibri
- Biology and Molecular Research Unit, Department of Life Sciences, National Center for Energy, Nuclear Science and Technology (CNESTEN), Rabat, Morocco
| | - Hassan Sefrioui
- Moroccan Foundation for Science, Innovation & Research (MAScIR), Centre de Biotechnologie Médicale, Rabat, Morocco
| | - Khalid Bougrin
- Equipe de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Faculty of Science, B.P. 1014, Geophysics, Natural Patrimony and Green Chemistry (GEOPAC) Research Center, Mohammed V University in Rabat, Rabat, Morocco.,Chemical and Biochemical Sciences Green Process Engineering (CBS), Mohammed VI Polytechnic University (UM6P), Benguerir, Morocco
| | - My El Abbes Faouzi
- Laboratory of Pharmacology and Toxicology, Biopharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
14
|
Ezati M, Ghavamipour F, Adibi H, Pouraghajan K, Arab SS, Sajedi RH, Khodarahmi R. Design, synthesis, spectroscopic characterizations, antidiabetic, in silico and kinetic evaluation of novel curcumin-fused aldohexoses. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 285:121806. [PMID: 36108405 DOI: 10.1016/j.saa.2022.121806] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/21/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
Curcumin (bis-α,β-unsaturated β-diketone) plays an important role in the prevention of numerous diseases, including diabetes. Curcumin, as an enzyme inhibitor, has ideal structural properties including hydrophobic nature, flexible backbone, and several available hydrogen bond (H-bond) donors and acceptors. In this study, curcumin-fused aldohexose derivatives 3(a-c) were synthesized and used as influential agents in the treatment of diabetes with inhibitory properties against two carbohydrate-hydrolyzing enzymes α-glucosidase (α-Gls) and α-amylase (α-Amy) which are known to be significant therapeutic targets for the reduction of postprandial hyperglycemia. These compounds were isolated, purified, and then spectrally characterized via FT-IR, Mass, 1H, and 13C NMR, which strongly confirmed the targeted product's formation. Also, their inhibitory properties against α-Gls and α-Amy were evaluated spectroscopically. The Results indicated that all compounds strongly inhibited α-Amy and α-Gls by mixed and competitive mechanisms, respectively. The intrinsic fluorescence of α-Amy was quenched by the interaction with compounds 1 and 3b through a dynamic quenching mechanism, and the 1 and 3b/α-Amy complexes were spontaneously formed, mainly driven by the hydrophobic interaction and hydrogen bonding. Fourier transform infrared spectra (FT-IR) comprehensively verified that the binding of compounds 1 and 3b to α-Amy would change the conformation and microenvironment of α-Amy, thereby inhibiting the enzyme activity. Docking and molecular dynamics (MD) simulations showed that all compounds interacted with amino acid residues located in the active pocket site of the proteins. In vivo studies confirmed the plasma glucose diminution after the administration of compound 3b to Wistar rats. Accordingly, the results of the current work may prompt the scientific communities to investigate the possibility of compound 3b application in the clinic.
Collapse
Affiliation(s)
- Mohammad Ezati
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fahimeh Ghavamipour
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hadi Adibi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khadijeh Pouraghajan
- Bioinformatics Laboratory, Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
15
|
Nawaz M, Taha M, Qureshi F, Ullah N, Selvaraj M, Shahzad S, Chigurupati S, Abubshait SA, Ahmad T, Chinnam S, Hisaindee S. Synthesis, α-amylase and α-glucosidase inhibition and molecular docking studies of indazole derivatives. J Biomol Struct Dyn 2022; 40:10730-10740. [PMID: 34463216 DOI: 10.1080/07391102.2021.1947892] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Herein, we report the synthesis and inhibitory potential of indazole (Methyl 1H-indazole-4-carboxylate) derivatives (1-13) against α-amylase and α-glucosidase enzymes. The described derivatives demonstrated good inhibitory potential with IC50 values, ranging between 15.04 ± 0.05 to 76.70 ± 0.06 µM ± SEM for α-amylase and 16.99 ± 0.19 to 77.97 ± 0.19 µM ± SEM for α-glucosidase, respectively. In particular, compounds (8-10 and 12) displayed significant inhibitory activities against both the screened enzymes, with their inhibitory potential comparable to the standard acarbose (12.98 ± 0.03 and 12.79 ± 0.17 µM ± SEM, respectively). Additionally, the influence of different substituents on enzyme inhibition activities was assessed to study the structure activity relationships. Molecular docking simulations were performed to rationalize the binding of derivatives/compounds with enzymes. All the synthesized derivatives (1-13) were characterized with the aid of spectroscopic instruments such as 1H-NMR, 13C-NMR, HR-MS, elemental analysis and FTIR.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Faiza Qureshi
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Deanship of Scientific Research, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nisar Ullah
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, Saudi Arabia
| | - Manikandan Selvaraj
- School of Chemical Engineering, Monash University, Selangor Darul Ehsan, Malaysia
| | - Sumaira Shahzad
- School of Business Administration, College of International Education, Zhejiang Gongshang University, Hangzhou, China
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Samar A Abubshait
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Department of Chemistry, College of Science and Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Tauqir Ahmad
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, Saudi Arabia
| | - Sampath Chinnam
- Department of Chemistry, B.M.S. College of Engineering, Bengaluru, Karnataka, India
| | - Soleiman Hisaindee
- Chemistry Department, College of Science, United Arab Emirates University, Al-Ain, UAE
| |
Collapse
|
16
|
Ali Y, Ahmad F, Ullah MF, Haq NU, Haq MIU, Aziz A, Zouidi F, Khan MI, Eldin SM. Structural Evaluation and Conformational Dynamics of ZNF141T474I Mutation Provoking Postaxial Polydactyly Type A. Bioengineering (Basel) 2022; 9:bioengineering9120749. [PMID: 36550955 PMCID: PMC9774408 DOI: 10.3390/bioengineering9120749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/24/2022] [Indexed: 12/04/2022] Open
Abstract
Postaxial Polydactyly (PAP) is a congenital disorder of limb abnormalities characterized by posterior extra digits. Mutations in the N-terminal region of the Zinc finger protein 141 (ZNF141) gene were recently linked with PAP type A. Zinc finger proteins exhibit similarity at their N-terminal regions due to C2-H2 type Zinc finger domains, but their functional preferences vary significantly by the binding patterns of DNA. Methods: This study delineates the pathogenic association, miss-fold aggregation, and conformational paradigm of a missense variant (c.1420C > T; p.T474I) in ZNF141 gene segregating PAP through a molecular dynamics simulations approach. Results: In ZNF141 protein, helices play a crucial role by attaching three specific target DNA base pairs. In ZNF141T474I protein, H1, H3, and H6 helices attain more flexibility by acquiring loop conformation. The outward disposition of the proximal portion of H9-helix in mutant protein occurs due to the loss of prior beta-hairpins at the C terminal region of the C2-H2 domain. The loss of hydrogen bonds and exposure of hydrophobic residues to solvent and helices turning to loops cause dysfunction of ZNF141 protein. These significant changes in the stability and conformation of the mutant protein were validated using essential dynamics and cross-correlation maps, which revealed that upon point mutation, the overall motion of the proteins and the correlation between them were completely different, resulting in Postaxial polydactyly type A. Conclusions: This study provides molecular insights into the structural association of ZNF141 protein with PAP type A. Identification of active site residues and legends offers new therapeutic targets for ZNF141 protein. Further, it reiterates the functional importance of the last residue of a protein.
Collapse
Affiliation(s)
- Yasir Ali
- Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak 27200, Pakistan
- National Centre for Bioinformatics, Quaid-I-Azam University, Islamabad 44000, Pakistan
| | - Faisal Ahmad
- National Centre for Bioinformatics, Quaid-I-Azam University, Islamabad 44000, Pakistan
| | - Muhammad Farhat Ullah
- National Centre for Bioinformatics, Quaid-I-Azam University, Islamabad 44000, Pakistan
| | - Noor Ul Haq
- Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak 27200, Pakistan
| | - M. Inam Ul Haq
- Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak 27200, Pakistan
| | - Abdul Aziz
- Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak 27200, Pakistan
| | - Ferjeni Zouidi
- Biology Department, Faculty of Arts and Sciences of Muhayil Aseer, King Khalid University, Abha 62529, Saudi Arabia
| | - M. Ijaz Khan
- Department of Mathematics and Statistics, Riphah International University I-14, Islamabad 44000, Pakistan
- Department of Mechanical Engineering, Lebanese American University, Beirut 13-5053, Lebanon
- Correspondence: or
| | - Sayed M. Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| |
Collapse
|
17
|
Alam A, Ali M, Latif A, Rehman NU, Saher S, Zainab, Faryal, Khan A, Ullah S, Ullah O, Halim SA, Sani F, Al-Harrasi A, Ahmad M. Novel Bis-Schiff’s base derivatives of 4-nitroacetophenone as potent α-glucosidase agents: Design, synthesis and in silico approach. Bioorg Chem 2022; 128:106058. [DOI: 10.1016/j.bioorg.2022.106058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/01/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
|
18
|
Efficient one-pot synthesis of arylated pyrazole-fused pyran analogs: as leads to treating diabetes and Alzheimer's disease. Future Med Chem 2022; 14:1507-1526. [PMID: 36268762 DOI: 10.4155/fmc-2022-0103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: To discover novel lead molecules against diabetes, Alzheimer's disease and oxidative stress, a library of arylated pyrazole-fused pyran derivatives, 1-20, were synthesized in a one-pot reaction. Materials & methods:1H-NMR spectroscopic and electron ionization mass spectrometry techniques were used to characterize the synthetic hybrid molecules 1-20. Analogs were screened against four indispensable therapeutic targets, including α-amylase, α-glucosidase, acetylcholinesterase and butyrylcholinesterase enzymes. Results: Except for derivatives 17 and 18, all other compounds exhibited varying degrees of inhibitory activities against target enzymes. The kinetic studies revealed that the synthetic molecules followed a competitive-type mode of inhibition for α-amylase and acetylcholinesterase enzymes, as well as a non-competitive mode of inhibition for α-glucosidase and butyrylcholinesterase enzymes. In addition, molecular docking studies identified crucial binding interactions of ligands with the enzyme's active site. Conclusion: These molecules may serve as a potential drug candidate to cure diabetes, Alzheimer's disease and oxidative stress in the future.
Collapse
|
19
|
Synthesis and evaluation of aryl aminomethylene substituted barbiturates and thiobarbiturates as novel α-amylase inhibitors and radical scavengers. MONATSHEFTE FUR CHEMIE 2022. [DOI: 10.1007/s00706-022-02972-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Recent Strategies in Transition-Metal-Catalyzed Sequential C–H Activation/Annulation for One-Step Construction of Functionalized Indazole Derivatives. Molecules 2022; 27:molecules27154942. [PMID: 35956893 PMCID: PMC9370621 DOI: 10.3390/molecules27154942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Designing new synthetic strategies for indazoles is a prominent topic in contemporary research. The transition-metal-catalyzed C–H activation/annulation sequence has arisen as a favorable tool to construct functionalized indazole derivatives with improved tolerance in medicinal applications, functional flexibility, and structural complexity. In the current review article, we aim to outline and summarize the most common synthetic protocols to use in the synthesis of target indazoles via a transition-metal-catalyzed C–H activation/annulation sequence for the one-step synthesis of functionalized indazole derivatives. We categorized the text according to the metal salts used in the reactions. Some metal salts were used as catalysts, and others may have been used as oxidants and/or for the activation of precatalysts. The roles of some metal salts in the corresponding reaction mechanisms have not been identified. It can be expected that the current synopsis will provide accessible practical guidance to colleagues interested in the subject.
Collapse
|
21
|
Hameed S, Khan KM, Taslimi P, Salar U, Taskin-Tok T, Kisa D, Saleem F, Solangi M, Ahmed MHU, Rani K. Evaluation of synthetic 2-aryl quinoxaline derivatives as α-amylase, α-glucosidase, acetylcholinesterase, and butyrylcholinesterase inhibitors. Int J Biol Macromol 2022; 211:653-668. [PMID: 35568155 DOI: 10.1016/j.ijbiomac.2022.05.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 04/30/2022] [Accepted: 05/05/2022] [Indexed: 01/13/2023]
Abstract
Variety of 2-aryl quinoxaline derivatives 1-23 were synthesized in good yields, by reacting 1,2-phenylenediamine with varyingly substituted phenacyl bromides in the presence of pyridine catalyst. All molecules 1-23 were characterized by spectroscopic techniques and evaluated for their diverse biological potential against α-amylase (α-AMY), α-glucosidase (α-GLU), acetylcholinesterase (AChE), and butyrylcholinesterase (BChE) enzymes. Synthetic derivatives possess enhanced inhibitory potential against all enzymes at nanomolar concentrations. In particular, compound 14 was found much superior with IC50 = 294.35, 198.21, 17.04, and 21.46 nM against α-AMY, α-GLU, AChE, and BChE, respectively, as compared to standard inhibitors. Furthermore, selected potent compounds, including 3, 4, 8, 14, 15, 17, and 18, were subjected to molecular docking studies to decipher the binding energies and interactions of ligands (synthetic molecules) with all four target enzymes.
Collapse
Affiliation(s)
- Shehryar Hameed
- 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.
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74100 Bartin, Turkey
| | - Uzma Salar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Tugba Taskin-Tok
- Gaziantep University, Faculty of Arts and Sciences, Department of Chemistry, Gaziantep, Turkey; Gaziantep University, Institute of Health Sciences, Department of Bioinformatics and Computational Biology, Gaziantep, Turkey
| | - Dursun Kisa
- Department of Molecular Biology and Genetics, Faculty of Science, Bartin University, 74100 Bartin, Turkey
| | - Faiza Saleem
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Mehwish Solangi
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Hassaan Uddin Ahmed
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Kiran Rani
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
22
|
Ke JJ, Lin J, Zhang X, Wu XZ, Zheng YY, Hu CM, Kang Y, Zhang K, Xiong Z, Ma ZQ. Synthesis of Benzylidene Analogs of Oleanolic Acid as Potential α-Glucosidase and α-Amylase Inhibitors. Front Chem 2022; 10:911232. [PMID: 35755256 PMCID: PMC9213889 DOI: 10.3389/fchem.2022.911232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Abstract
A series of benzylidene analogs of oleanolic acid 4a∼4s were synthesized and assessed for their α-glucosidase and α-amylase inhibitory activities. The results presented that all synthesized analogs exhibited excellent-to-moderate inhibitory effects on α-glucosidase and α-amylase. Analog 4i showed the highest α-glucosidase inhibition (IC50: 0.40 μM), and analog 4o presented the strongest α-amylase inhibition (IC50: 9.59 μM). Inhibition kinetics results showed that analogs 4i and 4o were reversible and mixed-type inhibitors against α-glucosidase and α-amylase, respectively. Simulation docking results demonstrated the interaction between analogs and two enzymes. Moreover, analogs 4i and 4o showed a high level of safety against 3T3-L1 and HepG2 cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhuang Xiong
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Zhi-Qiang Ma
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| |
Collapse
|
23
|
Mugaranja KP, Kulal A. Investigation of effective natural inhibitors for starch hydrolysing enzymes from Simaroubaceae plants by molecular docking analysis and comparison with in-vitro studies. Heliyon 2022; 8:e09360. [PMID: 35600433 PMCID: PMC9118686 DOI: 10.1016/j.heliyon.2022.e09360] [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: 09/18/2021] [Revised: 11/22/2021] [Accepted: 04/27/2022] [Indexed: 11/25/2022] Open
Abstract
The present study aims to find the effective natural enzyme inhibitors against alpha-amylase and alpha-glucosidase from the array of compounds identified in plants of the Simaroubaceae family using molecular docking and ADME/Toxicity studies. Among the 218 compounds docked against seven enzymes, buddlenol-A and citrusin-B showed the best binding energies (kcal/mol) of -7.830 and -7.383 against human salivary alpha-amylase and pancreatic alpha-amylase respectively. The other two compounds 9-hydroxycanthin-6-one and bruceolline-B had the best binding energy of -6.461 and -7.576 against N-terminal and C-terminal maltase glucoamylase respectively. Whereas the binding energy of prosopine (-6.499) and fisetinidol (-7.575) was considered as the best against N-terminal and C-terminal sucrase-isomaltase respectively. Picrasidine-X showed the best binding energy (-7.592) against yeast alpha-glucosidase. The study revealed that the seven compounds which showed the best binding energy against respective enzymes are considered as the 'lead hit compounds'. Even though the 'lead hit compounds' are not obeying all the laws of ADMET, the drug-likeness properties of 9-hydroxycanthin-6-one, fisetinidol, picrasidine-X, and prosopine were considerable. Also, kaempferol-3-O-pentoside was the recent compound identified from the Simarouba glauca plant extract found to be one among the top five lead hit compounds against four enzymes. This study provides valuable insight into the direction of developing natural compounds as potential starch hydrolysing enzyme inhibitors for managing type 2 diabetes.
Collapse
Affiliation(s)
- Kirana P. Mugaranja
- Biological Sciences Division, Poornaprajna Institute of Scientific Research, Bidalur Post, Devanahalli, Bangalore Rural, 562110, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - Ananda Kulal
- Biological Sciences Division, Poornaprajna Institute of Scientific Research, Bidalur Post, Devanahalli, Bangalore Rural, 562110, India
| |
Collapse
|
24
|
Solangi M, Kanwal, Khan KM, Chigurupati S, Saleem F, Qureshi U, Ul-Haq Z, Jabeen A, Felemban SG, Zafar F, Perveen S, Taha M, Bhatia S. Isatin thiazoles as antidiabetic: Synthesis, in vitro enzyme inhibitory activities, kinetics, and in silico studies. Arch Pharm (Weinheim) 2022; 355:e2100481. [PMID: 35355329 DOI: 10.1002/ardp.202100481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 11/11/2022]
Abstract
Diabetes mellitus is one of the most prevalent diseases nowadays. Several marketed drugs are available for the cure and treatment of diabetes, but there is still a dire need of introducing compatible drug molecules with lesser side effects. The current study is based on the synthesis of isatin thiazole derivatives 4-30 via the Hantzsch reaction. The synthetic compounds were characterized using different spectroscopic techniques and evaluated for their α-amylase and α-glucosidase inhibition potential. Of 27 isatin thiazoles, five (4, 5, 10, 12, and 16) displayed good activities against the α-amylase enzyme with IC50 values in the range of 22.22 ± 0.02-27.01 ± 0.06 µM, and for α-glucosidase, the IC50 values of these compounds were in the range of 20.76 ± 0.17-27.76 ± 0.17 µM, respectively. The binding interactions of the active molecules within the active site of enzymes were studied with the help of molecular docking studies. In addition, kinetic studies were carried out to examine the mechanism of action of the synthetic molecules as well. Compounds 3a, 4, 5, 10, 12, and 16 were also examined for their cytotoxic effect and were found to be noncytotoxic. Thus, several molecules were identified as good antihyperglycemic agents, which can be further modified to enhance inhibition ability and to find the lead molecule that can act as a potential antidiabetic agent.
Collapse
Affiliation(s)
- Mehwish Solangi
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, Pakistan
| | - Kanwal
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, Pakistan.,Institute of Marine Biotechnology, Universiti Malaysia Terengannu, Kuala Terengganu, Terengganu, Malaysia
| | - Khalid M Khan
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, Pakistan.,Department of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, Qassim University, Buraydah, Saudi Arabia
| | - Faiza Saleem
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, Pakistan
| | - Urooj Qureshi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Almas Jabeen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shatha G Felemban
- Department of Medical Laboratory Science, Fakeeh College for Medical Sciences, Jeddah, Kingdom of Saudi Arabia
| | - Fatima Zafar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Saurabh Bhatia
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| |
Collapse
|
25
|
Li J, Zhu J, Wu H, Li W. Synthesis, in vitro, and in silico studies of fisetin and quercetin and their metal complexes as inhibitors of α-glucosidase and thrombin. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
26
|
Maliwal D, Pissurlenkar RRS, Telvekar V. Identification of Novel Potential Anti-Diabetic Candidates targeting Human Pancreatic ɑ-Amylase and Human ɑ-Glycosidase: An Exhaustive Structure-based Screening. CAN J CHEM 2021. [DOI: 10.1139/cjc-2021-0238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetes is a major health issue that half a billion people affected worldwide. It is a serious, long-term medical condition majorly impacting the lives and well-being of individuals, families, and societies at large. It is amongst the top 10 diseases responsible for the death amongst adults with an expected rise to 10.2% (578 million) by 2030 and 10.9% (700 million) by 2045. The carbohydrates absorbed into the body are hydrolyzed by pancreatic α-amylase and other enzymes, human α-glucosidase. The α-amylase and α-glucosidase are validated therapeutic targets in the treatment of Type II diabetes (T2DM) as they play a vital role in modulating the blood glucose post meal. Herein, we report novel and diverse molecules as potential candidates, with predicted affinity for α-amylase and α-glucosidase. These molecules have been identified via hierarchical multistep docking of small molecules database with the estimated binding free energies. A Glide XP Score cutoff −8.00 kcal/mol was implemented to filter out non potential molecules. Four molecules viz. amb22034702, amb18105639, amb17153304, and amb9760832 have been identified after an exhaustive computational study involving, evaluation of binding interactions and assessment of the pharmacokinetics and toxicity profiles. The in-depth analysis of protein– ligand interactions was performed using a 100ns molecular dynamics (MD) simulation to establish the dynamic stability. Furthermore MM-GBSA based binding free energies were computed for 1000 trajectory snapshots to ascertain the strong binding affinity of these molecules for α-amylase and αglucosidase. The identified molecules can be considered as promising candidates for further drug development through necessary experimental assessments.
Collapse
Affiliation(s)
- Deepika Maliwal
- Institute of Chemical Technology, 52735, Department of Pharmaceutical Sciences and Technology, Mumbai, Maharashtra, India
| | | | - Vikas Telvekar
- Institute of Chemical Technology, 52735, Department of Pharmaceutical Sciences and Technology, Mumbai, Maharashtra, India
| |
Collapse
|
27
|
Saleem F, Kanwal, Mohammed Khan K, Chigurupati S, Andriani Y, Solangi M, Hameed S, Abdel Monem Abdel Hafez A, Begum F, Arif Lodhi M, Taha M, Rahim F, Sifzizul bin Tengku Muhammad T, Perveen S. Dicyanoanilines as potential and dual inhibitors of α-amylase and α-glucosidase enzymes: Synthesis, characterization, in vitro, in silico, and kinetics studies. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103651] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
28
|
Soto-Ospina A, Araque Marín P, Bedoya GDJ, Villegas Lanau A. Structural Predictive Model of Presenilin-2 Protein and Analysis of Structural Effects of Familial Alzheimer's Disease Mutations. Biochem Res Int 2021; 2021:9542038. [PMID: 34881055 PMCID: PMC8648483 DOI: 10.1155/2021/9542038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease manifests itself in brain tissue by neuronal death, due to aggregation of β-amyloid, produced by senile plaques, and hyperphosphorylation of the tau protein, which produces neurofibrillary tangles. One of the genetic markers of the disease is the gene that translates the presenilin-2 protein, which has mutations that favor the appearance of the disease and has no reported crystallographic structure. In view of this, protein modeling is performed using prediction and structural refinement tools followed by an energetic and stereochemical characterization for its validation. For the simulation, four reported mutations are chosen, which are Met239Ile, Met239Val, Ser130Leu, and Thr122Arg, all associated with various functional responses. From a theoretical analysis, a preliminary bioinformatic study is made to find the phosphorylation patterns in the protein and the hydropathic index according to the polarity and chemical environment. Molecular visualization was carried out with the Chimera 1.14 software, and the theoretical calculation with the hybrid quantum mechanics/molecular mechanics system from the semi-empirical method, with Spartan18 software and an AustinModel1 basis. These relationships allow for studying the system from a structural approach with the determination of small distance changes, potential surfaces, electrostatic maps, and angle changes, which favor the comparison between wild-type and mutant systems. With the results obtained, it is expected to complement experimental data reported in the literature from models that would allow us to understand the effects of the selected mutations.
Collapse
Affiliation(s)
- Alejandro Soto-Ospina
- University of Antioquia, Faculty of Medicine, Group Molecular Genetics, Medellín, Colombia
- University of Antioquia, Faculty of Medicine, Group Neuroscience of Antioquia, Medellín, Colombia
| | - Pedronel Araque Marín
- EIA University, School of Life Sciences, Research and Innovation in Chemistry Formulations Group, Envigado, Colombia
| | | | - Andrés Villegas Lanau
- University of Antioquia, Faculty of Medicine, Group Molecular Genetics, Medellín, Colombia
- University of Antioquia, Faculty of Medicine, Group Neuroscience of Antioquia, Medellín, Colombia
| |
Collapse
|
29
|
Saeedi M, Raeisi-Nafchi M, Sobhani S, Mirfazli SS, Zardkanlou M, Mojtabavi S, Faramarzi MA, Akbarzadeh T. Synthesis of 4-alkylaminoimidazo[1,2-a]pyridines linked to carbamate moiety as potent α-glucosidase inhibitors. Mol Divers 2021; 25:2399-2409. [PMID: 33047276 DOI: 10.1007/s11030-020-10137-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 08/25/2020] [Indexed: 02/05/2023]
Abstract
In this work, various imidazo[1,2-a]pyridines linked to carbamate moiety were designed, synthesized, and evaluated for their α-glucosidase inhibitory activity. Among synthesized compounds, 4-(3-(tert-Butylamino)imidazo[1,2-a]pyridin-2-yl)phenyl p-tolylcarbamate (6d) was the most potent compound (IC50 = 75.6 µM) compared with acarbose as the reference drug (IC50 = 750.0 µM). Kinetic study of compound 6d indicated a competitive inhibition. Also, the molecular docking study suggested desired interactions with the active site residues. In particular, hydrogen bonds and electrostatic interactions constructed by compound 6d afforded well-oriented conformation in the 3A4A active site.
Collapse
Affiliation(s)
- Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Raeisi-Nafchi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Sobhani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Sara Mirfazli
- Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Zardkanlou
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, 1417614411, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, 1417614411, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, 1417614411, Iran
| | - Tahmineh Akbarzadeh
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Mech D, Kurowska A, Trotsko N. The Bioactivity of Thiazolidin-4-Ones: A Short Review of the Most Recent Studies. Int J Mol Sci 2021; 22:11533. [PMID: 34768964 PMCID: PMC8584074 DOI: 10.3390/ijms222111533] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 01/28/2023] Open
Abstract
Thiazolidin-4-ones is an important heterocyclic ring system of a pharmacophore and a privileged scaffold in medicinal chemistry. This review is focused on the latest scientific reports regarding biological activities of thiazolidin-4-ones published in 2020 and 2021. The review covers recent information about antioxidant, anticancer, anti-inflammatory, analgesic, anticonvulsant, antidiabetic, antiparasitic, antimicrobial, antitubercular and antiviral properties of thiazolidin-4-ones. Additionally, the influence of different substituents in molecules on their biological activity was discussed in this paper. Thus, this study may help to optimize the structure of thiazolidin-4-one derivatives as more efficient drug agents. Presented information may be used as a practical hint for rational design of new small molecules with biological activity, especially among thiazolidin-4-ones.
Collapse
Affiliation(s)
| | | | - Nazar Trotsko
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (D.M.); (A.K.)
| |
Collapse
|
31
|
Mukhtar A, Shah S, Kanwal, Khan KM, Khan SU, Zaib S, Iqbal J, Parveen S, Taha M, Hussain S, Hameed S, Khan NA, Siddiqui R, Anwar A. Synthesis of Chalcones as Potential
α
‐Glucosidase Inhibitors,
In‐Vitro
and
In‐Silico
Studies. ChemistrySelect 2021. [DOI: 10.1002/slct.202102434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Asma Mukhtar
- H. E. J. Research Institute of Chemistry International Center for Chemical and Biological Sciences University of Karachi Karachi 75270 Pakistan
| | - Shazia Shah
- H. E. J. Research Institute of Chemistry International Center for Chemical and Biological Sciences University of Karachi Karachi 75270 Pakistan
| | - Kanwal
- H. E. J. Research Institute of Chemistry International Center for Chemical and Biological Sciences University of Karachi Karachi 75270 Pakistan
- Institute of Marine Biotechnology Universiti Malaysia Terengganu 21030 Kuala Terengganu Malaysia
| | - 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 1982 Dammam 31441 Saudi Arabia
| | - Shahid Ullah Khan
- Centre for Advanced Drug Research COMSATS University Islamabad Abbottabad Campus Abbottabad 22060 Pakistan
| | - Sumera Zaib
- Centre for Advanced Drug Research COMSATS University Islamabad Abbottabad Campus Abbottabad 22060 Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research COMSATS University Islamabad Abbottabad Campus Abbottabad 22060 Pakistan
| | - Shahnaz Parveen
- PCSIR Laboratories Complex Karachi, Shahra-e-Dr. Salimuzzaman Siddiqui Karachi 75280 Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy Institute for Research and Medical Consultations (IRMC) Imam Abdulrahman Bin Faisal University P.O. Box 1982 Dammam 31441 Saudi Arabia
| | - Shafqat Hussain
- Department of Chemistry University of Baltistan, Skardu, Main Campus Husainabad Kargil Road, Skardu Gilgit-Baltistan Pakistan
| | - Shahryar Hameed
- H. E. J. Research Institute of Chemistry International Center for Chemical and Biological Sciences University of Karachi Karachi 75270 Pakistan
| | - Naveed Ahmed Khan
- Department of Clinical Sciences College of Medicine University of Sharjah Sharjah 27272 United Arab Emirates
| | - Ruqaiyyah Siddiqui
- College of Arts and Sciences American University of Sharjah Sharjah 26666 United Arab Emirates
| | - Ayaz Anwar
- Department of Biological Sciences School of Medical and Life Sciences Sunway University Petaling Jaya 47500 Selangor Malaysia
| |
Collapse
|
32
|
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: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Zheng PF, Xiong Z, Liao CY, Zhang X, Feng M, Wu XZ, Lin J, Lei LS, Zhang YC, Wang SH, Xu XT. In vitro and in silico studies of bis (indol-3-yl) methane derivatives as potential α-glucosidase and α-amylase inhibitors. J Enzyme Inhib Med Chem 2021; 36:1938-1951. [PMID: 34459690 PMCID: PMC8409970 DOI: 10.1080/14756366.2021.1971976] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
In this paper, bis (indol-3-yl) methanes (BIMs) were synthesised and evaluated for their inhibitory activity against α-glucosidase and α-amylase. All synthesised compounds showed potential α-glucosidase and α-amylase inhibitory activities. Compounds 5 g (IC50: 7.54 ± 1.10 μM), 5e (IC50: 9.00 ± 0.97 μM), and 5 h (IC50: 9.57 ± 0.62 μM) presented strongest inhibitory activities against α-glucosidase, that were ∼ 30 times stronger than acarbose. Compounds 5 g (IC50: 32.18 ± 1.66 µM), 5 h (IC50: 31.47 ± 1.42 µM), and 5 s (IC50: 30.91 ± 0.86 µM) showed strongest inhibitory activities towards α-amylase, ∼ 2.5 times stronger than acarbose. The mechanisms and docking simulation of the compounds were also studied. Compounds 5 g and 5 h exhibited bifunctional inhibitory activity against these two enzymes. Furthermore, compounds showed no toxicity against 3T3-L1 cells and HepG2 cells.Highlights A series of bis (indol-3-yl) methanes (BIMs) were synthesised and evaluated inhibitory activities against α-glucosidase and α-amylase. Compound 5g exhibited promising activity (IC50 = 7.54 ± 1.10 μM) against α-glucosidase. Compound 5s exhibited promising activity (IC50 = 30.91 ± 0.86 μM) against α-amylase. In silico studies were performed to confirm the binding interactions of synthetic compounds with the enzyme active site.
Collapse
Affiliation(s)
- Peng-Fei Zheng
- Second Hospital of Lanzhou University, Lanzhou, PR China
| | - Zhuang Xiong
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| | - Cui-Ying Liao
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| | - Xin Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| | - Mei Feng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| | - Xiao-Zheng Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| | - Jing Lin
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| | - Lin-Sheng Lei
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, PR China
| | | | - Shao-Hua Wang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, PR China
| | - Xue-Tao Xu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, PR China
| |
Collapse
|
34
|
Hu CM, Wang WJ, Ye YN, Kang Y, Lin J, Wu PP, Li DL, Bai LP, Xu XT, Li BQ, Zhang K. Novel cinnamic acid magnolol derivatives as potent α-glucosidase and α-amylase inhibitors: Synthesis, in vitro and in silico studies. Bioorg Chem 2021; 116:105291. [PMID: 34438122 DOI: 10.1016/j.bioorg.2021.105291] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 01/06/2023]
Abstract
In this study, twenty novel cinnamic acid magnolol derivatives were synthesized, and screened for their anti-hyperglycemic potential. All synthesized compounds exhibited good to moderate α-glucosidase and α-amylase inhibitory activities with IC50 values: 5.11 ± 1.46-90.26 ± 1.85 µM and 4.27 ± 1.51-49.28 ± 2.54 µM as compared to the standard acarbose (IC50: 255.44 ± 1.89 μM and 80.33 ± 2.95 μM, respectively). Compound 6j showed the strongest inhibitory activity against α-glucosidase (IC50 = 5.11 ± 1.46 µM) and α-amylase (IC50 = 4.27 ± 1.51 µM). Kinetic study indicated that compound 6j was reversible and a mixed type inhibitor against α-glucosidase and α-amylase. In silico studies revealed the binding interaction between 6j and two enzymes, respectively. Finally, cells cytotoxicity assay revealed that compound 6j showed low toxicity against 3 T3-L1 cells and HepG2 cells.
Collapse
Affiliation(s)
- Chun-Mei Hu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Wen-Jing Wang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Yuan-Na Ye
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Yu Kang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Jing Lin
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Pan-Pan Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Dong-Li Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Li-Ping Bai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, PR China
| | - Xue-Tao Xu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China.
| | - Bao-Qiong Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China.
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China; School of biomedicine and pharmaceutical sciences, Guangdong University of Technology, Guangdong 510006, PR China.
| |
Collapse
|
35
|
Novel pyrano-triazolo-pyrimidine derivatives as anti- α-amylase agents: Synthesis, molecular docking investigations and computational analysis. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
36
|
Saeedi M, Eslami A, Mirfazli SS, Zardkanlou M, Faramarzi MA, Mahdavi M, Akbarzadeh T. Design and Synthesis of Novel 5-Arylisoxazole-1,3,4-thiadiazole Hybrids as α-Glucosidase Inhibitors. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999201104125018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
α-Glucosidase inhibitors have occupied a significant position in the treatment
of type 2 diabetes. In this respect, the development of novel and efficient non-sugar-based
inhibitors is in high demand.
Objective:
Design and synthesis of new 5-arylisoxazole-1,3,4-thiadiazole hybrids possessing α-
glucosidase inhibitory activity were developed.
Methods:
Different derivatives were synthesized by the reaction of various 5-arylisoxazole-3-
carboxylic acids and ethyl 2-((5-amino-1,3,4-thiadiazol-2-yl)thio)acetate. Finally, they were evaluated
for their α-glucosidase inhibitory activity.
Results:
It was found that ethyl 2-((5-(5-(2-chlorophenyl)isoxazole-3-carboxamido)-1,3,4-thiadiazol-
2-yl)thio)acetate (5j) was the most potent compound (IC50 = 180.1 μM) compared with acarbose as
the reference drug (IC50 = 750.0 μM). Also, the kinetic study of 5j revealed a competitive inhibition
and docking study results indicated desired interactions of that compound with amino acid residues
located close to the active site of α-glucosidase.
Conclusion:
Good α-glucosidase inhibitory activity obtained by the title compounds introduced
them as an efficient scaffold, which merits to be considered in anti-diabetic drug discovery
developments.
Collapse
Affiliation(s)
- Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Eslami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Sara Mirfazli
- Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Zardkanlou
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Akbarzadeh
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Mphahlele MJ, Magwaza NM, Malindisa ST, Choong YS. Biological evaluation the 2-aryl-2,3-dihydrobenzodiazaborinin-4(1H)-ones as potential dual α-glucosidase and α-amylase inhibitors with antioxidant properties. Chem Biol Drug Des 2021; 98:234-247. [PMID: 34013660 DOI: 10.1111/cbdd.13893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/03/2021] [Accepted: 05/15/2021] [Indexed: 12/16/2022]
Abstract
The 2-aryl-2,3-dihydrobenzodiazaborinin-4(1H)-ones (azaborininone) were synthesized as analogues of the 2-arylquinazoline-4-ones and screened through enzymatic assay in vitro for inhibitory effect against α-glucosidase and α-amylase activities. These azaborininones exhibited moderate to good inhibitory effect against these enzymes compared to acarbose used as a reference standard. The results are supported by the enzyme-ligand interactions through kinetics (in vitro) and molecular docking (in silico) studies. The test compounds also exhibited significant antioxidant activity through the 2,2-diphenyl-1-picrylhydrazyl (DPPH) and nitric oxide (NO) free radical scavenging assays. These azaborininone derivatives exhibited no effect on the viability of the human lung cancer (A549) cell line after 24 hr and were also not toxic towards the Vero cells.
Collapse
Affiliation(s)
- Malose J Mphahlele
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Florida, South Africa
| | - Nontokozo M Magwaza
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Florida, South Africa
| | - Sibusiso T Malindisa
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida, South Africa
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
38
|
Soto-Ospina A, Araque Marín P, Bedoya G, Sepulveda-Falla D, Villegas Lanau A. Protein Predictive Modeling and Simulation of Mutations of Presenilin-1 Familial Alzheimer's Disease on the Orthosteric Site. Front Mol Biosci 2021; 8:649990. [PMID: 34150846 PMCID: PMC8206637 DOI: 10.3389/fmolb.2021.649990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/22/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease pathology is characterized by β-amyloid plaques and neurofibrillary tangles. Amyloid precursor protein is processed by β and γ secretase, resulting in the production of β-amyloid peptides with a length ranging from 38 to 43 amino acids. Presenilin 1 (PS1) is the catalytic unit of γ-secretase, and more than 200 PS1 pathogenic mutations have been identified as causative for Alzheimer's disease. A complete monocrystal structure of PS1 has not been determined so far due to the presence of two flexible domains. We have developed a complete structural model of PS1 using a computational approach with structure prediction software. Missing fragments Met1-Glut72 and Ser290-Glu375 were modeled and validated by their energetic and stereochemical characteristics. Then, with the complete structure of PS1, we defined that these fragments do not have a direct effect in the structure of the pore. Next, we used our hypothetical model for the analysis of the functional effects of PS1 mutations Ala246GLu, Leu248Pro, Leu248Arg, Leu250Val, Tyr256Ser, Ala260Val, and Val261Phe, localized in the catalytic pore. For this, we used a quantum mechanics/molecular mechanics (QM/MM) hybrid method, evaluating modifications in the topology, potential surface density, and electrostatic potential map of mutated PS1 proteins. We found that each mutation exerts changes resulting in structural modifications of the active site and in the shape of the pore. We suggest this as a valid approach for functional studies of PS1 in view of the possible impact in substrate processing and for the design of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Alejandro Soto-Ospina
- Faculty of Medicine, Group Molecular Genetics, University of Antioquia, Medellín, Colombia
- Faculty of Medicine, Group Neuroscience of Antioquia, University of Antioquia, Medellín, Colombia
| | - Pedronel Araque Marín
- School of Life Sciences, Research and Innovation in Chemistry Formulations Group, EIA University, Envigado, Colombia
| | - Gabriel Bedoya
- Faculty of Medicine, Group Molecular Genetics, University of Antioquia, Medellín, Colombia
| | - Diego Sepulveda-Falla
- Faculty of Medicine, Group Neuroscience of Antioquia, University of Antioquia, Medellín, Colombia
- Molecular Neuropathology of Alzheimer’s Disease, Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrés Villegas Lanau
- Faculty of Medicine, Group Molecular Genetics, University of Antioquia, Medellín, Colombia
- Faculty of Medicine, Group Neuroscience of Antioquia, University of Antioquia, Medellín, Colombia
| |
Collapse
|
39
|
Mphahlele MJ, Agbo EN, Choong YS. Synthesis, Structure, Carbohydrate Enzyme Inhibition, Antioxidant Activity, In Silico Drug-Receptor Interactions and Drug-Like Profiling of the 5-Styryl-2-Aminochalcone Hybrids. Molecules 2021; 26:2692. [PMID: 34064448 PMCID: PMC8125089 DOI: 10.3390/molecules26092692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/28/2022] Open
Abstract
The 2-amino-5-(3/4-fluorostyryl)acetophenones were prepared and reacted with benzaldehyde derivatives to afford the corresponding 5-styryl-2-aminochalcone hybrids. The trans geometry of the styryl and α,β-unsaturated carbonyl arms, and the presence of NH…O intramolecular hydrogen bond were validated using 1H-NMR and X-ray data. The 2-amino-5-styrylacetophenones and their 5-styryl-2-aminochalcone derivatives were screened in vitro for their capability to inhibit α-glucosidase and/or α-amylase activities. Their antioxidant properties were evaluated in vitro through the 2,2-diphenyl-1-picrylhydrazyl (DPPH) and nitric oxide (NO) free radical scavenging assays. Kinetic studies of the most active derivatives from each series against α-glucosidase and/or α-amylase activities have been performed supported by molecular docking studies to determine plausible protein-ligand interactions on a molecular level. The key aspects of the pharmacokinetics of these compounds, i.e., absorption, distribution, metabolism, and excretion have also been simulated at theoretical level. The most active compounds from each series, namely, 2a and 3e, were evaluated for cytotoxicity against the normal monkey kidney cells (Vero cells) and the adenocarcinomic human epithelial (A549) cell line to establish their safety profile at least in vitro.
Collapse
Affiliation(s)
- Malose J. Mphahlele
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa;
| | - Emmanuel Ndubuisi Agbo
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa;
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| |
Collapse
|
40
|
Qin G, Xu W, Liu J, Zhao L, Chen G. Purification, characterization and hypoglycemic activity of glycoproteins obtained from pea (Pisum sativum L.). FOOD SCIENCE AND HUMAN WELLNESS 2021. [DOI: 10.1016/j.fshw.2021.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
41
|
Fettach S, Thari FZ, Hafidi Z, Tachallait H, Karrouchi K, El Achouri M, Cherrah Y, Sefrioui H, Bougrin K, Faouzi MEA. Synthesis, α-glucosidase and α-amylase inhibitory activities, acute toxicity and molecular docking studies of thiazolidine-2,4-diones derivatives. J Biomol Struct Dyn 2021; 40:8340-8351. [PMID: 33847536 DOI: 10.1080/07391102.2021.1911854] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In the present study, a series of thiazolidine-2,4-diones derivatives (3a-3e) and (4a-4e) were synthesized and characterized by 1H NMR, 13C NMR and ESI-MS spectrometry. All compounds were screened for their α-glucosidase and α-amylase inhibitory activities. In vitro biological investigations revealed that most of compounds were active against α-glucosidase with IC50 values in the range of 43.85 ± 1.06 to 380.10 ± 1.02 µM, and α-amylase with IC50 in the range of 18.19 ± 0.11 to 208.10 ± 1.80 µM. Some of the tested compounds were found to be more potent inhibitors than the clinical drug Acarbose (IC50glucosidase = 97.12 ± 0.35 µM and IC50amylase = 2.97 ± 0.004 μM). The lead compounds were evaluated for their acute toxicity on Swiss mice and found to be completely non-toxic with LD > 2000 mg/kg BW. Furthermore, the Structure-activity relationship (SAR) and the binding interactions of all compounds with the active site of α-glucosidase and α-amylase were confirmed through molecular docking and stabilizing energy calculations. This study has identified the inhibitory potential a new class of synthesized thiazolidine-2,4-diones in controlling both hyperglycemia and type 2 diabetes mellitus. Furthermore, the theoretical binding mode of the target molecules was evaluated by molecular docking studies against the 3D Crystal Structure of human pancreatic α-amylase (PDB ID: 1B2Y) and α-glucosidase (PDB ID: 3W37)Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saad Fettach
- Laboratory of Pharmacology and Toxicology, Biopharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Fatima Zahra Thari
- Equipe de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Faculty of Science, Geophysics, Natural Patrimony and Green Chemistry (GEOPAC) Research Center, Mohammed V University in Rabat, Rabat, Morocco
| | - Zakaria Hafidi
- Laboratoire de Physico-Chimie des Matériaux Inorganiques et Organiques, Centre des Sciences des Matériaux, Ecole Normale Supérieure-Rabat, Mohammed V University in Rabat, Rabat, Morocco
| | - Hamza Tachallait
- Equipe de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Faculty of Science, Geophysics, Natural Patrimony and Green Chemistry (GEOPAC) Research Center, Mohammed V University in Rabat, Rabat, Morocco
| | - Khalid Karrouchi
- Laboratory of Analytical Chemistry and Bromatology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Mohammed El Achouri
- Laboratoire de Physico-Chimie des Matériaux Inorganiques et Organiques, Centre des Sciences des Matériaux, Ecole Normale Supérieure-Rabat, Mohammed V University in Rabat, Rabat, Morocco
| | - Yahia Cherrah
- Laboratory of Pharmacology and Toxicology, Biopharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Hassan Sefrioui
- Moroccan Foundation for Science, Innovation & Research (MAScIR), Centre de Biotechnologie Médicale, Rabat, Morocco
| | - Khalid Bougrin
- Equipe de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Faculty of Science, Geophysics, Natural Patrimony and Green Chemistry (GEOPAC) Research Center, Mohammed V University in Rabat, Rabat, Morocco.,Mohammed VI Polytechnic University, Benguerir, Morocco
| | - My El Abbes Faouzi
- Laboratory of Pharmacology and Toxicology, Biopharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
42
|
Farooq S, Haq IU, Ullah N. Synthesis, characterization and biological evaluation of N-Mannich base derivatives of 2-phenyl-2-imidazoline as potential antioxidants, enzyme inhibitors, antimicrobials, cytotoxic and anti-inflammatory agents. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
43
|
Babatunde O, Hameed S, Salar U, Chigurupati S, Wadood A, Rehman AU, Venugopal V, Khan KM, Taha M, Perveen S. Dihydroquinazolin-4(1H)-one derivatives as novel and potential leads for diabetic management. Mol Divers 2021; 26:849-868. [PMID: 33650031 DOI: 10.1007/s11030-021-10196-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 02/05/2021] [Indexed: 11/25/2022]
Abstract
A variety of dihydroquinazolin-4(1H)-one derivatives (1-37) were synthesized via "one-pot" three-component reaction scheme by treating aniline and different aromatic aldehydes with isatoic anhydride in the presence of acetic acid. Chemical structures of compounds were deduced by different spectroscopic techniques including EI-MS, HREI-MS, 1H-, and 13C-NMR. Compounds were subjected to α-amylase and α-glucosidase inhibitory activities. A number of derivatives exhibited significant to moderate inhibition potential against α-amylase (IC50 = 23.33 ± 0.02-88.65 ± 0.23 μM) and α-glucosidase (IC50 = 25.01 ± 0.12-89.99 ± 0.09 μM) enzymes, respectively. Results were compared with the standard acarbose (IC50 = 17.08 ± 0.07 μM for α-amylase and IC50 = 17.67 ± 0.09 μM for α-glucosidase). Structure-activity relationship (SAR) was rationalized by analyzing the substituents effects on inhibitory potential. Kinetic studies were implemented to find the mode of inhibition by compounds which revealed competitive inhibition for α-amylase and non-competitive inhibition for α-glucosidase. However, in silico study identified several important binding interactions of ligands (synthetic analogues) with the active site of both enzymes.
Collapse
Affiliation(s)
- Oluwatoyin Babatunde
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Department of Chemical Sciences, Ajayi Crowther University, Oyo, P.M.B 1066, Nigeria
| | - Shehryar Hameed
- H. E. J. Research Institute of Chemistry, 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
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, 52571, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, UCSS, Abdul Wali Khan University, Mardan, Pakistan
| | - Ashfaq Ur Rehman
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, UCSS, Abdul Wali Khan University, Mardan, 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 1982, Dammam, 31441, Saudi Arabia.
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Karachi, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi, 75280, Pakistan
| |
Collapse
|
44
|
Chaudhry F, Shahid W, Al-Rashida M, Ashraf M, Ali Munawar M, Ain Khan M. Synthesis of imidazole-pyrazole conjugates bearing aryl spacer and exploring their enzyme inhibition potentials. Bioorg Chem 2021; 108:104686. [PMID: 33581666 DOI: 10.1016/j.bioorg.2021.104686] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Developing improved enzyme inhibitors is an effective therapy to counter various diseases. Aiming to build up biologically active templates, a new series of bis-diazoles conjugated with an aryl linker was designed and prepared through a convenient synthetic approach. Synthesized derivatives 6(a-m), having different substitutions at the 2nd position of the imidazole nucleus, depict the scope of present study. These compounds were characterized through spectroscopic methods and further examined for their in vitro enzyme inhibitory potentials against two selected enzymes: α-glucosidase and lipoxygenase (LOX). Overall, this series was found to be effective against α-glucosidase and moderately active against LOX enzyme. Compound 6k was the most potent α-glucosidase inhibitor with IC50 = 54.25 ± 0.67 µM as compared to reference drug acarbose (IC50 = 375.82 ± 1.76 µM). The docked conformation revealed the involvement of substituent's heteroatoms with amino acid residue Gly280 through hydrogen bonding. The most active LOX inhibitor was 6a with IC50 = 41.75 ± 0.04 µM as compared to standard baicalein (IC50 = 22.4 ± 1.3 µM). Docking model of 6a suggested the strong interaction of imidazole's nitrogen with iron atom of the active pocket of enzyme. Other features like lipophilicity, bulkiness of compounds, pi-pi interactions and/or pi-alkyl interactions also affected the inhibiting potentials of all prepared scaffolds.
Collapse
Affiliation(s)
- Faryal Chaudhry
- Institute of the Chemistry, Quaid-e-Azam Campus, University of the Punjab, Lahore 54590, Pakistan; Department of Chemistry, Kinnaird College for Women Lahore, 93-Jail Road, Lahore 54000, Pakistan.
| | - Wardah Shahid
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Ferozepur Road, Lahore 54600, Pakistan
| | - Muhammad Ashraf
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Munawar Ali Munawar
- Institute of the Chemistry, Quaid-e-Azam Campus, University of the Punjab, Lahore 54590, Pakistan.
| | - Misbahul Ain Khan
- Institute of the Chemistry, Quaid-e-Azam Campus, University of the Punjab, Lahore 54590, Pakistan; Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| |
Collapse
|
45
|
Synthesis, crystal structure, hirshfeld surface analysis, DFT calculations, anti-diabetic activity and molecular docking studies of (E)-N’-(5-bromo-2-hydroxybenzylidene) isonicotinohydrazide. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128800] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
46
|
Rafique R, Arshia, Kanwal, Khan KM, Chigurupati S, Salar U, Taha M, Perveen S. Discovery of New N-hydrazinecarbothioamide Indazole Hybrids: As Potential Radical (ABTS and DPPH) Scavengers. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817999200424074455] [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
Background:
Free radicals are the main cause of numerous diseases. Their overproduction
needs to be controlled in order to combat several ailments. The current study deals with the discovery
of new free radical scavengers.
Methods:
Substituted N-hydrazinecarbothioamide indazoles 1-18 were evaluated for DPPH and
ABTS radical scavenging activities.
Results:
All synthetic compounds possess good radical DPPH and ABTS scavenging
potential in the ranges of IC50 = 2.11 ± 0.17 - 5.3 ± 0.11 μM and IC50 = 2.31 ± 0.06 - 5.5 ± 0.07
μM, respectively, as compared to standard ascorbic acid having IC50 = 2.02 ± 0.11 μM for DPPH
and IC50 = 2.1 ± 0.07 μM for ABTS.
Conclusion:
These compounds could serve as leads for antioxidant activity that have the ability to
control free radical generation and ward off free radical-induced disorders.
Collapse
Affiliation(s)
- Rafaila Rafique
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Arshia
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Kanwal
- 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
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - 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, Dammam, P.O. Box 31441, Saudi Arabia
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi 75280, Pakistan
| |
Collapse
|
47
|
Szmigiel-Bakalarz K, Nentwig M, Günther D, Oeckler O, Malik-Gajewska M, Michalska D, Morzyk-Ociepa B. Sodium and lithium one-dimensional coordination polymers with 1H-indazole-3-carboxylic acid: Crystal structures, vibrational spectra and DFT calculations. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Shamim S, Khan KM, Ullah N, Chigurupati S, Wadood A, Ur Rehman A, Ali M, Salar U, Alhowail A, Taha M, Perveen S. Synthesis and screening of (E)-3-(2-benzylidenehydrazinyl)-5,6-diphenyl-1,2,4-triazine analogs as novel dual inhibitors of α-amylase and α-glucosidase. Bioorg Chem 2020; 101:103979. [PMID: 32544738 DOI: 10.1016/j.bioorg.2020.103979] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/16/2020] [Accepted: 05/27/2020] [Indexed: 01/08/2023]
Abstract
(E)-3-(2-Benzylidenehydrazinyl)-5,6-diphenyl-1,2,4-triazines analogs 1-27 were synthesized by multi-step reaction scheme and subjected to in vitro inhibitory screening against α-amylase and α-glucosidase enzymes. Out of these twenty-seven synthetic analogs, ten compounds 14-17, 19, and 21-25 are structurally new. All compounds exhibited good to moderate inhibitory potential in terms of IC50 values ranging (IC50 = 13.02 ± 0.04-46.90 ± 0.05 µM) and (IC50 = 13.09 ± 0.08-46.44 ± 0.24 µM) in comparison to standard acarbose (IC50 = 12.94 ± 0.27 µM and 10.95 ± 0.08 µM), for α-amylase and α-glucosidase, respectively. Structure-activity relationship indicated that analogs with halogen substitution(s) were found more active as compared to compounds bearing other substituents. Kinetic studies on most active α-amylase and α-glucosidase inhibitors 5, 7, 9, 15, 24, and 27, suggested non-competitive and competitive types of inhibition mechanism for α-amylase and α-glucosidase, respectively. Molecular docking studies predicted the good protein-ligand interaction (PLI) profile with key interactions such as arene-arene, H-<, <-<, and <-H etc., against the corresponding targets.
Collapse
Affiliation(s)
- Shahbaz Shamim
- 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.
| | - Nisar Ullah
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran-31261, Saudi Arabia
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Shankar Campus, Abdul Wali Khan University, Mardan, Khyber Pukhtoonkhwa, Pakistan
| | - Ashfaq Ur Rehman
- Department of Biochemistry, Shankar Campus, Abdul Wali Khan University, Mardan, Khyber Pukhtoonkhwa, Pakistan
| | - Muhammad Ali
- H. E. J. Research Institute of Chemistry, 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
| | - Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia
| | - 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
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Karachi, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi-75280, Pakistan
| |
Collapse
|
49
|
Novel Azoles as Antiparasitic Remedies against Brain-Eating Amoebae. Antibiotics (Basel) 2020; 9:antibiotics9040188. [PMID: 32316387 PMCID: PMC7235764 DOI: 10.3390/antibiotics9040188] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Balamuthia mandrillaris and Naegleria fowleri are opportunistic protozoan pathogens capable of producing infection of the central nervous system with more than 95% mortality rate. Previously, we have synthesized several compounds with antiamoebic properties; however, synthesis of compounds that are analogues of clinically used drugs is a highly desirable approach that can lead to effective drug development against these devastating infections. In this regard, compounds belonging to the azole class possess wide range of antimicrobial properties and used clinically. In this study, six novel benzimidazole, indazole, and tetrazole derivatives were synthesized and tested against brain-eating amoebae. These compounds were tested for their amoebicidal and static properties against N. fowleri and B. mandrillaris. Furthermore, the compounds were conjugated with silver nanoparticles and characterized. The synthetic heterocyclic compounds showed up to 72% and 65% amoebicidal activities against N. fowleri and B. mandrillaris respectively, while expressing up to 75% and 70% amoebistatic activities, respectively. Following conjugation with silver nanoparticles, amoebicidal activities of the drugs increased by up to 46 and 36% versus B. mandrillaris and N. fowleri. Minimal effects were observed when the compounds were evaluated against human cells using cytotoxicity assays. In summary, azole compounds exhibited potent activity against N. fowleri and B. mandrillaris. Moreover, conjugation of the azole compounds with silver nanoparticles further augmented the capabilities of the compounds against amoebae.
Collapse
|
50
|
Kawde AN, Taha M, Alansari RS, Almandil NB, Anouar EH, Uddin N, Rahim F, Chigurupati S, Nawaz M, Hayat S, Ibrahim M, Elakurthy PK, Vijayan V, Morsy M, Ibrahim H, Baig N, Khan KM. Exploring efficacy of indole-based dual inhibitors for α-glucosidase and α-amylase enzymes: In silico, biochemical and kinetic studies. Int J Biol Macromol 2020; 154:217-232. [PMID: 32173438 DOI: 10.1016/j.ijbiomac.2020.03.090] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 01/25/2023]
Abstract
α-Glucosidase and α-amylase are enzymes which are associated with diabetic II. These enzymes break macromolecules of sugar into monosugar molecules which is soluble in body, hence increase the sugar level in blood. There is need to develop economical and save inhibitors to prevent them from breaking sugar macromolecules to soluble molecules which will control the level of sugar in blood. Therefore, we synthesized indole-based derivatives (1-18) and evaluated as dual inhibitor for α-glucosidase and α-amylase. These chemical scaffolds were built with variation in aryl ring which were found active with good to moderate activity for α-glucosidase having IC50 value ranging from 13.99 ± 0.10 to 59.09 ± 0.30 μM when compared with standard acarbose with IC50 of 11.29 ± 0.10 μM; for α-amylase IC50 value ranging from 13.14 ± 0.10 to 58.99 ± 0.30 μM when compared with the standard acarbose with IC50 of 11.12 ± 0.10 μM. Structure activity relationship (SAR) has been established for all compounds. Enzymatic kinetic study and molecular docking study have been carried out to investigate the binding interactions α-glucosidase and α-amylase enzyme.
Collapse
Affiliation(s)
- Abdel-Nasser Kawde
- Chemistry Department, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia.
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia.
| | - Raneem Saud Alansari
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Noor Barak Almandil
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - El Hassane Anouar
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Nizam Uddin
- Department of Chemistry, University of Karachi, Karachi 75270, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21300, Khyber Pakhtunkhwa, Pakistan
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Shawkat Hayat
- Department of Chemistry, Hazara University, Mansehra 21300, Khyber Pakhtunkhwa, Pakistan
| | - Mohamad Ibrahim
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | | | | | - Mohamed Morsy
- Chemistry Department, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Hossieny Ibrahim
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Nadeem Baig
- Chemistry Department, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|