1
|
Hariri R, Saeedi M, Mojtabavi S, Alizadeh S, Ebadi A, Faramarzi MA, Amini M, Sharifzadeh M, Biglar M, Akbarzadeh T. Design, synthesis, and investigation of novel 5-arylpyrazole-glucose hybrids as α-glucosidase inhibitors. Sci Rep 2025; 15:9912. [PMID: 40121215 PMCID: PMC11929827 DOI: 10.1038/s41598-025-92706-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Considering the global incidence of diabetes, developing new compounds to lower blood sugar levels has become increasingly crucial. As a result, there has been a growing focus on the synthesis of α-glucosidase inhibitors in recent years. This study investigated design, synthesis, and effects of novel 5-aryl pyrazole-glucose hybrids as α-glucosidase inhibitors. Thirteen derivatives from this class of compounds were synthesized, demonstrating superior in vitro inhibitory effects (IC50 values ranging from 0.5 to 438.6 µM, compared to acarbose at 750.0 µM). Among them, compound 8g (IC50 = 0.5 µM) was selected for further investigations and the kinetic studies revealed that it is a competitive inhibitor (Ki = 0.46 µM). Fluorescence assays indicated changes in the fluorescence intensity, while thermodynamic analyses suggested that compound 8g promoted a transition of the enzyme into an unfolded state. Furthermore, in vivo studies demonstrated that 8g effectively reduced blood sugar levels in rats at doses comparable to acarbose. Molecular docking studies revealed that this compound interacted with the enzyme's active site, and molecular dynamics simulations showed that pharmacophores engaged in various interactions with the enzyme.
Collapse
Affiliation(s)
- Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - 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
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Simin Alizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ahmad Ebadi
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Biglar
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Chen YT, Wan BW, Wang KM, Zhu KK, Meng N, Jiang CS, Zhang J. Design and synthesis of N-(3-cyanothiophen-2-yl)-2-phenoxyacetamide-based α-glucosidase inhibitors. Bioorg Med Chem Lett 2025; 117:130068. [PMID: 39662707 DOI: 10.1016/j.bmcl.2024.130068] [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: 10/11/2024] [Revised: 11/17/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
This study investigates the design and synthesis of a series of novel selective α-glucosidase inhibitors based on N-(3-cyanothiophen-2-yl)-2-phenoxyacetamide framework, employing a bioisosterism strategy. Among the nineteen newly synthesized analogs, compound 4d9 demonstrated the highest α-glucosidase inhibitory potency (IC50 = 2.11 μM) when compared to the established inhibitors Acarbose (IC50 = 327.0 μM) and HXH8r (IC50 = 15.32 μM), while exhibiting a remarkable 17.48-fold selectivity for α-glucosidase over α-amylase. Kinetic studies revealed that compound 4d9 acts as a non-competitive inhibitor, and its binding interactions were further investigated using molecular docking analysis. Additionally, compound 4d9 showed noncytotoxic effects on human normal hepatocyte (LO2) cells and demonstrated improved metabolic stability in rat plasma. These findings position compound 4d9 as a promising candidate for the development of therapeutics targeting type 2 diabetes.
Collapse
Affiliation(s)
- Yi-Tong Chen
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Bo-Wen Wan
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kai-Ming Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kong-Kai Zhu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
3
|
Singh R, Sindhu J, Singh D, Kumar P. Key molecular scaffolds in the development of clinically viable α-amylase inhibitors. Future Med Chem 2025; 17:347-362. [PMID: 39835704 PMCID: PMC11792802 DOI: 10.1080/17568919.2025.2453421] [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: 10/27/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
The escalating cases of type II diabetes combined with adverse side effects of current antidiabetic drugs spurred the advancement of innovative approaches for the management of postprandial glucose levels. α-Amylase is an endoamylase responsible for the breakdown of internal α-1,4-glycosidic linkages in dietary starch, producing oligosaccharides. Subsequently, α-glucosidase degraded these oligosaccharides to monosaccharides, which are absorbed into the bloodstream and become available to the body. The inhibitors of α-amylase reduced the digestibility of carbohydrates accompanied by delayed glucose absorption, leading to decreased blood glucose levels after meals and thus, inhibition of the enzyme seems to be a crucial strategy for diabetes management and improving overall glycemic control in diabetic patients. The present review article emphasizes the therapeutic promise of recently discovered potential α-amylase inhibitors, highlighting their in vitro, in silico and in vivo profiles. Ultimately, we addressed the contemporary challenges and potential routes ahead in the search for safe and reliable α-amylase inhibitors for clinical use, summarizing the most recent research in the field.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
- School of Chemistry, Indian Institutes of Science Education and Research, Thiruvananthapuram, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
4
|
Hosseini Nasab N, Raza H, Eom YS, Shah FH, Kwak JH, Kim SJ. Exploring chalcone-sulfonyl piperazine hybrids as anti-diabetes candidates: design, synthesis, biological evaluation, and molecular docking study. Mol Divers 2025; 29:43-59. [PMID: 38775996 DOI: 10.1007/s11030-024-10831-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/25/2024] [Indexed: 02/02/2025]
Abstract
To address the escalating rates of diabetes mellitus worldwide, there is a growing need for novel compounds. The demand for more affordable and efficient methods of managing diabetes is increasing due to the inevitable side effects associated with existing antidiabetic medications. In this present research, various chalcone-sulfonyl piperazine hybrid compounds (5a-k) were designed and synthesized to develop inhibitors against alpha-glucosidase and alpha-amylase. In addition, several spectroscopic methods, including FT-IR, 1H-NMR, 13C-NMR, and HRMS, were employed to confirm the exact structures of the synthesized derivatives. All synthesized compounds were evaluated for their ability to inhibit alpha-glucosidase and alpha-amylase in vitro using acarbose as the reference standard and they showed excellent to good inhibitory potentials. Compound 5k exhibited excellent inhibitory activity against alpha-glucosidase (IC50 = 0.31 ± 0.01 µM) and alpha-amylase (IC50 = 4.51 ± 1.15 µM), which is 27-fold more active against alpha-glucosidase and 7-fold more active against alpha-amylase compared to acarbose, which had IC50 values of 8.62 ± 1.66 µM for alpha-glucosidase and 30.97 ± 2.91 µM for alpha-amylase. It was discovered from the Lineweaver-Burk plot that 5k exhibited competitive inhibition against alpha-glucosidase. Furthermore, cytotoxicity screening assay results against human fibroblast HT1080 cells showed that all compounds had a good level of safety profile. To explore the binding interactions of the most potent compound (5k) with the active site of enzymes, molecular docking research was conducted, and the results obtained supported the experimental data.
Collapse
Affiliation(s)
- Narges Hosseini Nasab
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Hussain Raza
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Young Seok Eom
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Fahad Hassan Shah
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Jae-Hwan Kwak
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea.
| |
Collapse
|
5
|
Tong J, Yan J, Zhang Y, Xing X. Novel α-glucosidase Inhibitors Designed as Type 2 Diabetes Drugs by QSAR, Molecular Docking and Molecular Dynamics Simulation Methods. Chem Biodivers 2025; 22:e202401674. [PMID: 39271631 DOI: 10.1002/cbdv.202401674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Diabetes mellitus is a globally prevalent disease of significant concern. Alpha-glucosidase has emerged as a prominent target for the treatment of type 2 diabetes. In this study, 39 α-glucosidase inhibitors (AGIs) of tetrahydrobenzo[b]thiophene-2-ylurea derivatives to establish a stable and valid Topomer CoMFA model, with a cross-validation coefficient (q2) of 0.766 and a non-cross-validation coefficient (r2) of 0.960. Subsequently, the ZINC15 database was used to screen the fragments, based on which 13 novel inhibitor molecules with theoretically potentially high activity were designed. Molecular docking and molecular dynamics simulations to understand the binding status of the inhibitor molecules to the target proteins showed that amino acids ASP215, GLN279 and ARG442 may form hydrogen bonds with the ligands and therefore enhance the inhibitory effect of the small molecules. Additionally, MM/PBSA calculations indicate that the newly designed molecules exhibit more stable binding modes. These molecules also demonstrate favorable ADMET properties with potential as AGIs. The findings would provide valuable guidance and a theoretical foundation for the design and development of novel AGIs.
Collapse
Affiliation(s)
- Jianbo Tong
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Xi'an, 710021, China
| | - Jing Yan
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Xi'an, 710021, China
| | - Yakun Zhang
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Xi'an, 710021, China
| | - Xiaoyu Xing
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Xi'an, 710021, China
| |
Collapse
|
6
|
Vadabingi N, Taneja AK, Mallepogu V, Pasala C, Meriga B, Amineni UM, Ponne CV, Sri KN, Tej MB, Mallapu RE. Design, Synthesis, and Biological Evaluation of Novel Bisurea Derivatives of p-Xylylenediamine as Potent Anti-Diabetic Agents. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2024; 60:S95-S111. [DOI: 10.1134/s107042802413013x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 03/14/2025]
|
7
|
Thakur S, Kumar D, Jaiswal S, Goel KK, Rawat P, Srivastava V, Dhiman S, Jadhav HR, Dwivedi AR. Medicinal chemistry-based perspectives on thiophene and its derivatives: exploring structural insights to discover plausible druggable leads. RSC Med Chem 2024:d4md00450g. [PMID: 39601022 PMCID: PMC11588141 DOI: 10.1039/d4md00450g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024] Open
Abstract
Thiophene is a privileged pharmacophore in medicinal chemistry owing to its diversified biological attributes. The thiophene moiety has been ranked 4th in the US FDA drug approval of small drug molecules, with around 7 drug approvals over the last decade. The present review covers USFDA-approved drugs possessing a thiophene ring system. Our analysis reveals that 26 drugs possessing thiophene nuclei have been approved under different pharmacological classes. The review further covers reported thiophene and its substituted analogues with diverse biological activities, including anti-diabetic, anticancer, anti-inflammatory, anticonvulsant, and antioxidant activity. Besides, a section is dedicated to appreciating the implications of structural bioinformatics in drug discovery. Additionally, the manuscript delves into structure-activity relationship studies to explore the chemical groups responsible for eliciting potential therapeutic activities. The review may provide invaluable insights for researchers working with thiophene nuclei in developing novel analogues with greater efficacy and fewer side effects.
Collapse
Affiliation(s)
- Shikha Thakur
- Pharmaceutical Chemistry Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani - 333031 RJ India
| | - Devendra Kumar
- School of Pharmacy, Narsee Monjee Institute of Management Studies (NMIMS) Dist. Dhule Maharashtra India
| | - Shivani Jaiswal
- Institute of Pharmaceutical Research, GLA University Mathura, 17 Km Stone, National Highway, Delhi-Mathura Road, P.O. Chaumuha Mathura-281406 Uttar Pradesh India
| | - Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University) Haridwar 249404 Uttarakhand India
| | - Pramod Rawat
- Graphic Era (Deemed to be University) Clement Town Dehradun-248002 India
- Graphic Era Hill University Clement Town Dehradun-248002 India
| | - Vivek Srivastava
- Amity Institute of Pharmacy Amity University Lucknow Campus Uttar Pradesh India
| | - Sonia Dhiman
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University Rajpura 140401 Punjab India
| | - Hemant R Jadhav
- Pharmaceutical Chemistry Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani - 333031 RJ India
| | - Ashish Ranjan Dwivedi
- Department of Medicinal Chemistry, GITAM School of Pharmacy, GITAM (deemed to be) University Hyderabad India
| |
Collapse
|
8
|
Song YZ, Zhang J, Song QJ, Zhu WH, Yuan C, Wang KM, Jiang CS. Synthesis and biological evaluation of novel 1,2,3,4-tetrahydro-β-carboline derivatives as potential antibacterial agents. Bioorg Med Chem Lett 2024; 109:129822. [PMID: 38823728 DOI: 10.1016/j.bmcl.2024.129822] [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: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
The quest for novel antibacterial agents is imperative in the face of escalating antibiotic resistance. Naturally occurring tetrahydro-β-carboline (THβC) alkaloids have been highlighted due to their significant biological derivatives. However, these structures have been little explored for antibacterial drugs development. In this study, a series of 1,2,3,4-THβC derivatives were synthesized and assessed for their antibacterial prowess against both gram-positive and gram-negative bacteria. The compounds exhibited moderate to good antibacterial activity, with some compounds showing superior efficacy against gram-positive bacteria, especially methicillin-resistant Staphylococcus aureus (MRSA), to that of Gentamicin. Among these analogs, compound 3k emerged as a hit compound, demonstrating rapid bactericidal action and a significant post-antibacterial effect, with significant cytotoxicity towards human LO2 and HepG2 cells. In addition, compound 3k (10 mg/kg) showed comparable anti-MRSA efficacy to Ciprofloxacin (2 mg/kg) in a mouse model of abdominal infection. Overall, the present findings suggested that THβC derivatives based on the title compounds hold promising applications in the development of antibacterial drugs.
Collapse
Affiliation(s)
- Yuan-Ze Song
- Zibo Hospital of Traditional Chinese Medicine, Zibo 255000, China
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Qing-Jiang Song
- Zibo Hospital of Traditional Chinese Medicine, Zibo 255000, China
| | - Wen-Hao Zhu
- Zibo Hospital of Traditional Chinese Medicine, Zibo 255000, China
| | - Chao Yuan
- Zoucheng Administration for Market Regulation, Zoucheng 273100, China
| | - Kai-Ming Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
9
|
Ullah S, Ullah A, Waqas M, Halim SA, Pasha AR, Shafiq Z, Mali SN, Jawarkar RD, Khan A, Khalid A, Abdalla AN, Kashtoh H, Al-Harrasi A. Structural, dynamic behaviour, in-vitro and computational investigations of Schiff's bases of 1,3-diphenyl urea derivatives against SARS-CoV-2 spike protein. Sci Rep 2024; 14:12588. [PMID: 38822113 PMCID: PMC11143201 DOI: 10.1038/s41598-024-63345-9] [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: 12/15/2023] [Accepted: 05/28/2024] [Indexed: 06/02/2024] Open
Abstract
The COVID-19 has had a significant influence on people's lives across the world. The viral genome has undergone numerous unanticipated changes that have given rise to new varieties, raising alarm on a global scale. Bioactive phytochemicals derived from nature and synthetic sources possess lot of potential as pathogenic virus inhibitors. The goal of the recent study is to report new inhibitors of Schiff bases of 1,3-dipheny urea derivatives against SARS COV-2 spike protein through in-vitro and in-silico approach. Total 14 compounds were evaluated, surprisingly, all the compounds showed strong inhibition with inhibitory values between 79.60% and 96.00% inhibition. Here, compounds 3a (96.00%), 3d (89.60%), 3e (84.30%), 3f (86.20%), 3g (88.30%), 3h (86.80%), 3k (82.10%), 3l (90.10%), 3m (93.49%), 3n (85.64%), and 3o (81.79%) exhibited high inhibitory potential against SARS COV-2 spike protein. While 3c also showed significant inhibitory potential with 79.60% inhibition. The molecular docking of these compounds revealed excellent fitting of molecules in the spike protein receptor binding domain (RBD) with good interactions with the key residues of RBD and docking scores ranging from - 4.73 to - 5.60 kcal/mol. Furthermore, molecular dynamics simulation for 150 ns indicated a strong stability of a complex 3a:6MOJ. These findings obtained from the in-vitro and in-silico study reflect higher potency of the Schiff bases of 1,3-diphenyl urea derivatives. Furthermore, also highlight their medicinal importance for the treatment of SARS COV-2 infection. Therefore, these small molecules could be a possible drug candidate.
Collapse
Affiliation(s)
- Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Atta Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Anam Rubbab Pasha
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Suraj N Mali
- School of Pharmacy, D.Y. Patil University (Deemed to be University), Sector 7, Nerul, Navi Mumbai, 400706, India
| | - Rahul D Jawarkar
- Department of Medicinal Chemistry and Drug Discovery, Dr. Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, 444603, India
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, 45142, Jazan, Saudi Arabia
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, 21955, Makkah, Saudi Arabia
| | - Hamdy Kashtoh
- Department of Biotechnology, Yeungnam University, Gyeongsan, 38541, Gyeongbuk, Republic of Korea.
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman.
| |
Collapse
|
10
|
Naseem S, Fatima S, Ullah S, Khan A, Mali SN, Jawarkar RD, Syed A, Elgorban AM, Al-Harrasi A, Shafiq Z. Carbonylbis(hydrazine-1-carbothioamide) derivatives as a new class of α-glucosidase inhibitors and their mechanistic insights via molecular docking and dynamic simulations. Arch Pharm (Weinheim) 2024; 357:e2300604. [PMID: 38148299 DOI: 10.1002/ardp.202300604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/28/2023]
Abstract
In the past, efforts have been made to find a cure for diabetes, mainly evaluating new classes of compounds to explore their potency. In this study, we present the synthesis and evaluation of carbonylbis(hydrazine-1-carbothioamide) derivatives as potential α-glucosidase inhibitors, employing both in vivo and in silico investigations. The in vitro experiments revealed that all tested compounds were significantly potent for α-glucosidase inhibition, with the lead compound 3a displaying approximately 80 times higher activity than acarbose. To delve deeper, in silico induced fit docking, pharmacokinetics, and molecular dynamics studies were conducted. Significantly, compound 3a exhibited a docking score of -7.87 kcal/mol, surpassing acarbose, which had a docking score of -6.59 kcal/mol. The in silico ADMET indicated that most of the synthesized compounds have properties conducive to drug development. Molecular dynamics analysis demonstrated that, when the ligand 3a was coupled with the target 3TOP, Cα-RMSD backbone RMSD values below 2.4 Å and "Lig_fit_Prot" values below 2.7 Å were observed. QSAR analysis demonstrates that the "fOC8A" descriptor positively correlates with α-glucosidase inhibition activity, while "lipoplus_AbSA" positively contributes and "notringC_notringO_8B" negatively contributes to this activity.
Collapse
Affiliation(s)
- Saira Naseem
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, Pakistan
| | - Shamool Fatima
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saeed Ullah
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Suraj N Mali
- Department of Pharmaceutical Science and Technology, Birla Institute of Technology, Mesra, India
| | - Rahul D Jawarkar
- Department of Medicinal Chemistry and Drug Discovery, Dr. Rajendra Gode Institute of Pharmacy, Amravati, India
| | - Asad Syed
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Zahid Shafiq
- Department of Pharmaceutical & Medicinal Chemistry, Bonn, Germany
| |
Collapse
|
11
|
Pathak S, Singh AP, Sharma R, Pandey R. An Overview of the Pharmacological Activities and Synthesis of Benzothiophene Derivatives. Med Chem 2024; 20:839-854. [PMID: 38920062 DOI: 10.2174/0115734064315107240603055845] [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: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024]
Abstract
One important class of organic compounds having many uses, especially in medical chemistry, is benzothiophene and its derivatives. This review examines the biological activity of benzothiophene derivatives and summarizes the synthetic methods used in their production. The effectiveness of several synthetic pathways, such as cyclization techniques, functional group modifications, and reactions catalyzed by transition metals, in gaining access to benzothiophene scaffolds has been examined. Additionally, a broad spectrum of therapeutic domains, such as antiinflammatory, antibacterial, antidiabetic, anticancer, antimicrobial, anti-leishmanial, antifungal, antimalarial, and antitubercular activities, are covered by the pharmacological activities that are being explored. The synthesis and pharmacological potential of benzothiophene derivatives are well-explained in this thorough review, which opens up new options for medicinal chemistry and drug discovery study. Overall, this study is a useful resource for scientists working on drug development and discovery as it sheds light on the pharmacological potential of benzothiophene derivatives. This review includes the synthesis and bioactivities of the years 2002-2024. The goal of this review is to compile the existing information on benzothiophene derivatives and provide guidance for future research and development as well as insights into their possible medicinal uses.
Collapse
Affiliation(s)
- Shilpi Pathak
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Ansh Pratap Singh
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Richa Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Rahul Pandey
- Department of Management, Dr. D. Y. Patil Arts, Commerce & Science College, Pune, India
| |
Collapse
|
12
|
Luo S, Yang W, Huang Y, Peng Z, Wang G. Design, synthesis, biological evaluation, and docking study of new triazole-phenylacetamide derivatives as α-glucosidase inhibitors. Bioorg Chem 2023; 141:106844. [PMID: 37703743 DOI: 10.1016/j.bioorg.2023.106844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/26/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
To discover potent α-glucosidase inhibitors, a class of novel triazole-phenylacetamide derivatives (5a-5p) were designed, prepared, and tested for their α-glucosidase inhibitory effects. All tested compounds (5a-5p) displayed a strong α-glucosidase inhibitory activity (IC50 = 6.69 ± 0.18-113.65 ± 2.94 μM) in comparison with the positive control acarbose (IC50 = 723.06 ± 11.26 μM). Thereinto, 5g (IC50 = 6.69 ± 0.18 μM) showed the best anti-α-glucosidase activity and behaved as a mixed-type inhibitor with the value of Ki and Kis to be 1.65 μM and 4.54 μM, respectively. Besides, fluorescence quenching experiment, three-dimensional fluorescence spectra assay, circular dichroism analysis, and molecular docking studies indicated that 5g may inhibit α-glucosidase activity by binding with its active site as well as changing the secondary structure of α-glucosidase. Combined with the inhibition effect on the rise of postprandial blood glucose level and low cytotoxicity of 5g, it could be concluded that these title compounds may play a role as lead compounds to develop novel α-glucosidase inhibitors.
Collapse
Affiliation(s)
- Shuang Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China; School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Wei Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China; School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yong Huang
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Zhiyun Peng
- Clinical Trails Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Guangcheng Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
13
|
Khan Y, Khan S, Hussain R, Maalik A, Rehman W, Attwa MW, Masood R, Darwish HW, Ghabbour HA. The Synthesis, In Vitro Bio-Evaluation, and In Silico Molecular Docking Studies of Pyrazoline-Thiazole Hybrid Analogues as Promising Anti-α-Glucosidase and Anti-Urease Agents. Pharmaceuticals (Basel) 2023; 16:1650. [PMID: 38139777 PMCID: PMC10747725 DOI: 10.3390/ph16121650] [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: 10/21/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
In the present work, a concise library of benzothiazole-derived pyrazoline-based thiazole (1-17) was designed and synthesized by employing a multistep reaction strategy. The newly synthesized compounds were screened for their α-glucosidase and urease inhibitory activities. The scaffolds (1-17) were characterized using a combination of several spectroscopic techniques, including FT-IR, 1H-NMR, 13C-NMR, and EI-MS. The majority of the synthesized compounds demonstrated a notable potency against α-glucosidase and urease enzymes. These analogues disclosed varying degrees of α-glucosidase and urease inhibitory activities, with their IC50 values ranging from 2.50 to 17.50 μM (α-glucosidase) and 14.30 to 41.50 (urease). Compounds 6, 7, 14, and 12, with IC50 values of 2.50, 3.20, 3.40, and 3.50 μM as compared to standard acarbose (IC50 = 5.30 µM), while the same compounds showed 14.30, 19.20, 21.80, and 22.30 comparable with thiourea (IC50 = 31.40 μM), respectively, showed excellent inhibitory activity. The structure-activity relationship revealed that the size and electron-donating or electron-withdrawing effects of substituents influenced the enzymatic activities such as α-glucosidase and urease. Compound 6 was a dual potent inhibitor against α-glucosidase and urease due to the presence of -CF3 electron-withdrawing functionality on the phenyl ring. To the best of our knowledge, these synthetic compounds were found to be the most potent dual inhibitors of α-glucosidase and urease with minimum IC50 values. Moreover, in silico studies on most active compounds, i.e., 6, 7, 14, and 12, were also performed to understand the binding interaction of most active compounds with active sites of α-glucosidase and urease enzymes.
Collapse
Affiliation(s)
- Yousaf Khan
- Department of Chemistry, COMSATS University Islamabad Campus, Islamabad 45550, Pakistan; (Y.K.); (R.M.)
| | - Shoaib Khan
- Department of Chemistry, Abbottabad University of Science and Technology (AUST), Abbottabad 22500, Pakistan;
| | - Rafaqat Hussain
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan;
| | - Aneela Maalik
- Department of Chemistry, COMSATS University Islamabad Campus, Islamabad 45550, Pakistan; (Y.K.); (R.M.)
| | - Wajid Rehman
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan;
| | - Mohamed W. Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.W.A.)
| | - Rafia Masood
- Department of Chemistry, COMSATS University Islamabad Campus, Islamabad 45550, Pakistan; (Y.K.); (R.M.)
| | - Hany W. Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.W.A.)
| | - Hazem A. Ghabbour
- School of Health and Biomedical Sciences, RMIT University, Melbourne 3083, Australia;
| |
Collapse
|
14
|
Elsebaie HA, El-Bastawissy EA, Elberembally KM, Khaleel EF, Badi RM, Shaldam MA, Eldehna WM, Tawfik HO, El-Moselhy TF. Novel 4-(2-arylidenehydrazineyl)thienopyrimidine derivatives as anticancer EGFR inhibitors: Design, synthesis, biological evaluation, kinome selectivity and in silico insights. Bioorg Chem 2023; 140:106799. [PMID: 37625210 DOI: 10.1016/j.bioorg.2023.106799] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
The current study discovered fifteen new thieno[2,3-d]pyrimidine derivatives with potential anticancer action, including 5a-l, 6, and 7a-b. Results from the NCI screening revealed that compounds 5f-i and 7a significantly inhibited the proliferation of MDA-MB-468 cells at mean GI% and GI50 levels. Compared to staurosporine, these compounds (5f-i and 7a) demonstrated better safety towards typical WI-38 cells. Compounds 5g and 7a demonstrated the highest inhibition (two-digit nanomolar) when compared to erlotinib when their potency was tested on EGFR kinase. Considering the outcomes above, 5g was examined for its ability to disrupt the cell cycle with trigger apoptosis in breast cancer MDA-MB-468 cell lines. The apoptosis markers Bax, Bcl-2, Caspase-8, and Caspase-9, were detected. In silico molecular docking and dynamic simulation were used to explainthe biological activities of the most potent compound.
Collapse
Affiliation(s)
- Heba A Elsebaie
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527 Egypt.
| | - Eman A El-Bastawissy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527 Egypt.
| | - Kamel M Elberembally
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527 Egypt.
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, King Khalid University, Asir 61421, Saudi Arabia.
| | - Rehab Mustafa Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, King Khalid University, Asir 61421, Saudi Arabia.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; School of Biotechnology, Badr University in Cairo, Badr City 11829, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527 Egypt.
| | - Tarek F El-Moselhy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527 Egypt.
| |
Collapse
|
15
|
Chahal S, Punia J, Rani P, Singh R, Mayank, Kumar P, Kataria R, Joshi G, Sindhu J. Development of thiazole-appended novel hydrazones as a new class of α-amylase inhibitors with anticancer assets: an in silico and in vitro approach. RSC Med Chem 2023; 14:757-781. [PMID: 37122544 PMCID: PMC10131644 DOI: 10.1039/d2md00431c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperamylasemia is reported to be associated with numerous chronic diseases, including diabetes and cancer. Considering this fact, we developed a series of thiazole-clubbed hydrazones. The derivatives were explored for their in vitro α-amylase inhibitory activity, which was further corroborated with their anticancer assets using a panel of cancer cells, including colon cancer (HCT-116), lung cancer (A549), and breast cancer (MDA-MB-231). To better understand pharmacokinetics, the synthetic derivatives were subjected to in silico ADMET prediction. The in vitro based biological investigation revealed that compared to the reference drug acarbose (IC50 = 0.21 ± 0.008 μM), all the synthesized compounds (5a-5aa) exhibited in vitro α-amylase inhibitory response in the range of IC50 values from 0.23 ± 0.003 to 0.5 ± 0.0 μM. Along with this, the proliferations of the HCT-116, A549 and MDA-MB-231 cells were inhibited when treated with the synthesized compounds. Notable cancer cell growth inhibition was observed for compounds 5e, 5f and 5y, which correlated with their α-amylase inhibition. Additionally, the kinetics investigation revealed that 5b, 5e, 5f and 5y exhibit uncompetitive inhibition. 5b was found to be the least cytotoxic and most potent α-amylase inhibitor and was further validated by absorption and fluorescence quenching technique.
Collapse
Affiliation(s)
- Sandhya Chahal
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Jyoti Punia
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Payal Rani
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Rajvir Singh
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| | - Mayank
- 3IT - Université de Sherbrooke 3000 Bd de l'Université Immeuble P2 Sherbrooke QC J1K 0A5 Canada
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University Kurukshetra 136119 India
| | - Ramesh Kataria
- Department of Chemistry, Panjab University Chandigarh 160014 India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal (A Central) University Chauras Campus, Tehri Garhwal 249161 Uttarakhand India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University Hisar 125004 India
| |
Collapse
|
16
|
Nguyen TH, Amen Y, Wang D, Othman A, Matsumoto M, Nagata M, Shimizu K. Oligomeric Proanthocyanidin Complex from Avocado Seed as A Promising α-glucosidase Inhibitor: Characteristics and Mechanisms. PLANTA MEDICA 2023; 89:316-323. [PMID: 35714650 DOI: 10.1055/a-1878-3916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Although considered an abundant source of agricultural by-products, avocado (Persea americana Mill.) seed, with several biological activities and bioactive components, might become a promising resource for phytopharmaceutical development. In this study, through bioassay-guided isolation of the main α-glucosidase inhibitors in avocado seed, we discovered the major α-glucosidase inhibitor to be avocado seed oligomeric proanthocyanidin complex (ASOPC). Thiolysis and UPLC-DAD-HRESIMS showed the presence of A- and B-type procyanidins, and B-type propelargonidin with (epi)afzelechin as extension unit. Mean degree of polymerization (mDP) of ASOPC was calculated as 7.3 ± 1. Furthermore, ASOPC appeared to be a strong, reversible, competitive inhibitor of α-glucosidase, with IC50 value of 0.1 µg/mL, which was significantly lower than Acarbose (IC50 = 75.6 µg/mL), indicated that ASOPC is a potential natural α-glucosidase inhibitor. These findings would contribute to the direction of utilizing avocado seed bioactive components with the possibility to be used as natural anti-diabetic agents.
Collapse
Affiliation(s)
- Thien Huu Nguyen
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
- Faculty of Chemical Engineering and Food Technology, Nong Lam University - Ho Chi Minh city, Vietnam
| | - Yhiya Amen
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dongmei Wang
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
| | - Ahmed Othman
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Masako Matsumoto
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
| | - Maki Nagata
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
| | - Kuniyoshi Shimizu
- Department of Agro-environmental Sciences, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University
| |
Collapse
|
17
|
Design, synthesis, biological evaluation, and docking study of chromone-based phenylhydrazone and benzoylhydrazone derivatives as antidiabetic agents targeting α-glucosidase. Bioorg Chem 2023; 132:106384. [PMID: 36696731 DOI: 10.1016/j.bioorg.2023.106384] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
To develop novel α-glucosidase inhibitors, a series of chromone-based phenylhydrazone and benzoylhydrazone derivatives were designed, synthesized, and evaluated their inhibitory effects on α-glucosidase. The target compounds were characterized using 1H NMR, 13C NMR, and high-resolution mass spectra. Some of the compounds showed a varying degree of α-glucosidase inhibitory activity with IC50 values ranging from 6.59 ± 0.09 to 158.55 ± 0.87 μM. Among them, compound 5c (IC50 = 6.59 ± 0.09 μM) was the most potent inhibitor by comparison with positive control acarbose (IC50 = 685.11 ± 7.46 μM). Enzyme kinetic, fluorescence analysis, circular dichroism spectra, and molecular docking techniques were employed to explain the underlying molecular mechanisms of 5c inhibition on α-glucosidase. In vivo sucrose-loading test showed that 5c could suppress the rise of blood glucose levels after loading sucrose in normal Kunming mice. The cytotoxicity assay indicated that 5c exhibited low cytotoxicity.
Collapse
|
18
|
Wu MJ, Yu DD, Du YQ, Zhang J, Su MZ, Jiang CS, Guo YW. Further undescribed cembranoids from South China Sea soft coral Sarcophyton ehrenbergi: Structural elucidation and biological evaluation. PHYTOCHEMISTRY 2023; 206:113549. [PMID: 36481314 DOI: 10.1016/j.phytochem.2022.113549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 06/17/2023]
Abstract
A detailed chemical investigation of the South China Sea soft coral Sarcophyton ehrenbergi has yield seven undescribed cembranoids, namely isoehrenbergol D and sarcoehrenolides F-K embodying a rare α,β-unsaturated-lactone moiety at C-6 to C-19, along with two known related compounds, ehrenbergol D and sarcoehrenolide A. Their structures and absolute configurations were unambiguously established in the light of extensive spectroscopic data analysis, modified Mosher's method, X-ray diffraction analysis, and quantum chemical computation method. In a bioassay for α-glucosidase inhibition, ehrenbergol D was evaluated as α-glucosidase inhibitor with an IC50 value of 13.57 μM.
Collapse
Affiliation(s)
- Meng-Jun Wu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals and College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Dan-Dan Yu
- College of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Ye-Qing Du
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Ming-Zhi Su
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Yue-Wei Guo
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals and College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China; College of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| |
Collapse
|
19
|
Pasha AR, Khan A, Ullah S, Halim SA, Hussain J, Khalid M, Naseer MM, El-Kott AF, Negm S, Al-Harrasi A, Shafiq Z. Synthesis of new diphenyl urea-clubbed imine analogs and its Implications in diabetic management through in vitro and in silico approaches. Sci Rep 2023; 13:1877. [PMID: 36725861 PMCID: PMC9892044 DOI: 10.1038/s41598-023-28828-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Type II diabetes mellitus (T2DM) is a global health issue with high rate of prevalence. The inhibition of α-glucosidase enzyme has prime importance in the management of T2DM. This study was established to synthesize Schiff bases of 1,3-dipheny urea (3a-y) and to investigate their in vitro anti-diabetic capability via inhibiting α-glucosidase, a key player in the catabolism of carbohydrates. The structures of all compounds were confirmed through various techniques including, Fourier-transform infrared spectroscopy (FTIR) and nuclear magnetic resonance (NMR) and mass-spectrometry (MS) methods. Interestingly all these compounds displayed potent inhibition IC50 values in range of 2.14-115 µM as compared to acarbose used as control. Additionally, all the compounds were docked at the active site of α-glucosidase to predict their mode of binding. The docking results indicates that Glu277 and Asn350 play important role in the stabilization of these compounds in the active site of enzyme. These molecules showed excellent predicted pharmacokinetics, physicochemical and drug-likeness profile. The anti-diabetic potential of these molecules signifies their medical importance and provide insights into prospective therapeutic options for the treatment of T2DM.
Collapse
Affiliation(s)
- Anam Rubbab Pasha
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan.,Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman.,International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Javid Hussain
- Department of Biological Sciences and Chemistry, University of Nizwa, Nizwa-616, Nizwa, Oman
| | - Muhammad Khalid
- Department of Chemistry, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, 64200, Pakistan.,Centre for Theoretical and Computational Research, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, 64200, Pakistan
| | | | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, 61421, Abha, Saudi Arabia.,Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Sally Negm
- Department of Life Sciences, College of Science and Art Mahyel Aseer, King Khalid University, 62529, Abha, Saudi Arabia.,Unit of Food Bacteriology, Central Laboratory of Food Hygiene, Ministry of Health, Branch in Zagazig, Zagazig, 44511, Egypt
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman.
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan. .,Department of Pharmaceutical and Medicinal Chemistry, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
20
|
Gao M, Ma H, Liu X, Zhang Y, Tang L, Zheng Z, Zhang X, Jiang C, Lin L, Sun H. Synthesis and Biological Evaluation of Substituted Pyrazole-Fused Oleanolic Acid Derivatives as Novel Selective α-Glucosidase Inhibitors. Chem Biodivers 2023; 20:e202201178. [PMID: 36573561 DOI: 10.1002/cbdv.202201178] [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: 12/12/2022] [Accepted: 12/27/2022] [Indexed: 12/28/2022]
Abstract
A series of novel substituted pyrazole-fused oleanolic acid derivative were synthesized and evaluated as selective α-glucosidase inhibitors. Among these analogs, compounds 4a-4f exhibited more potent inhibitory activities compared with their methyl ester derivatives, and standard drugs acarbose and miglitol as well. Besides, all these analogs exhibited good selectivity towards α-glucosidase over α-amylase. Analog 4d showed potent inhibitory activity against α-glucosidase (IC50 =2.64±0.13 μM), and greater selectivity towards α-glucosidase than α-amylase by ∼33-fold. Inhibition kinetics showed that compound 4d was a non-competitive α-glucosidase inhibitor, which was consistent with the result of its simulation molecular docking. Moreover, the in vitro cytotoxicity of compounds 4a-4f towards hepatic LO2 and HepG2 cells was tested.
Collapse
Affiliation(s)
- Mei Gao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.,Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Hui Ma
- Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Xiaoyu Liu
- Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Yanhua Zhang
- Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Liansheng Tang
- Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Zhiyong Zheng
- Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Xinlei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Chengshi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Lin Lin
- Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Haiji Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| |
Collapse
|
21
|
Wang KM, Ge YX, Zhang J, Chen YT, Zhang NY, Gu JS, Fang L, Zhang XL, Zhang J, Jiang CS. New cycloalkyl[b]thiophenylnicotinamide-based α-glucosidase inhibitors as promising anti-diabetic agents: Synthesis, in silico study, in vitro and in vivo evaluations. Bioorg Med Chem Lett 2023; 79:129069. [PMID: 36395995 DOI: 10.1016/j.bmcl.2022.129069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
In the present study, a series of cycloalkyl[b]thiophenylnicotinamide derivatives against α-glucosidase were synthesized, and evaluated for their in vitro and in vivo anti-diabetic potential. Most of the synthetic analogues exhibited superior α-glucosidase inhibitory effects than the standard drug acarbose (IC50 = 258.5 μM), in which compound 11b with cyclohexyl[b]thiophene core demonstrated the highest activity with an IC50 value of 9.9 μM and showed higher selectivity towards α-glucosidase over α-amylase by 7.4-fold. Fluorescence quenching experiment confirmed the direct binding of 11b with α-glucosidase, kinetics study revealed that 11b was a mixed-type inhibitor, and its binding mode was analyzed using molecular docking. Moreover, analogs compounds 6a-9b, 11b, 12b did not show in vitro cytotoxicity against LO2 and HepG2 cells. Finally, compound 11b exhibited in vivo hypoglycemic activity by reducing the blood glucose levels in sucrose-loaded rats.
Collapse
Affiliation(s)
- Kai-Ming Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yong-Xi Ge
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jie Zhang
- Shandong Boyuan Pharmaceutical & Chemical Co., Ltd., Shouguang 262725, China
| | - Yi-Tong Chen
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Nai-Yu Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Jin-Song Gu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Lei Fang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Xin-Lei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, Shaanxi, China.
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
22
|
Mohammadi‐Khanaposhtani M, Noori M, Valizadeh Y, Dastyafteh N, Ghomi MK, Mojtabavi S, Faramarzi MA, Hosseini S, Biglar M, Larijani B, Rastegar H, Hamedifar H, Mirzazadeh R, Mahdavi M. Synthesis, α‐glucosidase Inhibition,
in silico
Pharmacokinetic, and Docking Studies Of Thieno[2,3‐b]Quinoline‐Acetamide Derivatives as New Anti‐Diabetic Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202104482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Maryam Mohammadi‐Khanaposhtani
- Cellular and Molecular Biology Research Center Health Research Institute Babol University of Medical Sciences Babol Iran
| | - Milad Noori
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Yousef Valizadeh
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Navid Dastyafteh
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Minoo Khalili Ghomi
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
| | | | - Mahmood Biglar
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Hossein Rastegar
- Cosmetic products research center, Iranian food and drug administration, MOHE Tehran Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center Alborz University of Medical Sciences Karaj Iran
| | | | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
23
|
Design, synthesis, and in silico studies of quinoline-based-benzo[d]imidazole bearing different acetamide derivatives as potent α-glucosidase inhibitors. Sci Rep 2022; 12:14019. [PMID: 35982225 PMCID: PMC9386204 DOI: 10.1038/s41598-022-18455-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
In this study, 18 novel quinoline-based-benzo[d]imidazole derivatives were synthesized and screened for their α-glucosidase inhibitory potential. All compounds in the series except 9q showed a significant α-glucosidase inhibition with IC50 values in the range of 3.2 ± 0.3–185.0 ± 0.3 µM, as compared to the standard drug acarbose (IC50 = 750.0 ± 5.0 µM). A kinetic study indicated that compound 9d as the most potent derivative against α-glucosidase was a competitive type inhibitor. Furthermore, the molecular docking study revealed the effective binding interactions of 9d with the active site of the α-glucosidase enzyme. The results indicate that the designed compounds have the potential to be further studied as new anti-diabetic agents.
Collapse
|
24
|
Noori M, Rastak M, Halimi M, Ghomi MK, Mollazadeh M, Mohammadi-Khanaposhtani M, Sayahi MH, Rezaei Z, Mojtabavi S, Ali Faramarzi M, Larijani B, Biglar M, Amanlou M, Mahdavi M. Design, synthesis, in vitro, and in silico enzymatic evaluations of thieno[2,3-b]quinoline-hydrazones as novel inhibitors for α-glucosidase. Bioorg Chem 2022; 127:105996. [PMID: 35878449 DOI: 10.1016/j.bioorg.2022.105996] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022]
Abstract
In the development of novel anti-α-glucosidase agents, we synthesized novel thieno[2,3-b]quinoline-hydrazones 9a-n by facile and efficient conventional chemical reactions. These compounds were characterized by IR, 1H NMR, 13C NMR, and elemental analysis. Inhibitory activities of the title compounds were evaluated against yeast α-glucosidase. In particular, compounds 9c, 9d, and 9h exhibited high anti-α-glucosidase activity. Representatively, compound 9c with IC50 = 1.3 µM, was 576-times more potent than positive control acarbose. Molecular docking study of the most active compounds showed that these compounds formed important binding interactions at α-glucosidase active site. Molecular dynamics study of compound 9c was also performed and the obtained results were compared with acarbose. Compounds 9c, 9d, and 9h were also evaluated for in silico druglikeness properties and ADMET prediction. These studies showed that the title most potent compounds could be exploited as drug candidates.
Collapse
Affiliation(s)
- Milad Noori
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mryam Rastak
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Halimi
- Department of Biology, Babol Branch, Islamic Azad University, Babol, Iran
| | - Minoo Khalili Ghomi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mrjan Mollazadeh
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Hosein Sayahi
- Department of Chemistry, Payame Noor University (PNU), P.O. Box 19395-3697, Tehran, Iran
| | - Zahra Rezaei
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Chen SQ, Jia J, Hu JY, Wu J, Sun WT, Zheng M, Wang X, Zhu KK, Jiang CS, Yang SP, Zhang J, Wang SB, Cai YS. Iboga-type alkaloids with Indolizidino[8,7-b]Indole scaffold and bisindole alkaloids from Tabernaemontana bufalina Lour. PHYTOCHEMISTRY 2022; 196:113089. [PMID: 35074605 DOI: 10.1016/j.phytochem.2022.113089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/08/2022] [Accepted: 01/09/2022] [Indexed: 06/14/2023]
Abstract
Phytochemical investigation on the aerial parts of Tabernaemontana bufalina Lour. (Apocynaceae) led to the identification of four undescribed monoterpenoid indole alkaloids named taberbufamines A-D, an undescribed natural product, and fourteen known indole alkaloids. The structures of the undescribed alkaloids were established by spectroscopic and computational methods, and their absolute configurations were further determined by quantum chemical TDDFT calculations and the experimental ECD spectra. Taberbufamines A and B possessed an uncommon skeleton incorporating an indolizidino [8,7-b]indole motif with a 2-hydroxymethyl-butyl group attached at the pyrrolidine ring. Biosynthetically, Taberbufamines A and B might be derived from iboga-type alkaloid through rearrangement. Vobatensine C showed significant bioactivity against A-549, Bel-7402, and HCT-116 cells with IC50 values of 2.61, 1.19, and 1.74 μM, respectively. Ervahanine A showed antimicrobial activity against Bacillus subtilis, Mycobacterium smegmatis, and Helicobacter pylori with MIC values of 4, 8, and 16 μg/mL, respectively. 19(S)-hydroxyibogamine was shown as butyrylcholinesterase inhibitor (IC50 of 20.06 μM) and α-glycosidase inhibitor (IC50 of 17.18 μM), while tabernamine, ervahanine B, and ervadivaricatine B only showed α-glycosidase inhibitory activities with IC50 values in the range of 0.95-4.61 μM.
Collapse
Affiliation(s)
- Shun-Qing Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jia Jia
- Department of Pathogen Biology & Jiangsu Key Laboratory of Pathogen Biology & Helicobacter Pylori Research Centre, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Jing-Yao Hu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jun Wu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Wen-Ting Sun
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Mingxin Zheng
- Department of Pathogen Biology & Jiangsu Key Laboratory of Pathogen Biology & Helicobacter Pylori Research Centre, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Xi Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Kong-Kai Zhu
- College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, People's Republic of China
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, People's Republic of China
| | - Sheng-Ping Yang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, People's Republic of China.
| | - Shou-Bao Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China.
| | - You-Sheng Cai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
26
|
Mehmood R, Sadiq A, Alsantali RI, Mughal EU, Alsharif MA, Naeem N, Javid A, Al-Rooqi MM, Chaudhry GES, Ahmed SA. Synthesis and Evaluation of 1,3,5-Triaryl-2-Pyrazoline Derivatives as Potent Dual Inhibitors of Urease and α-Glucosidase Together with Their Cytotoxic, Molecular Modeling and Drug-Likeness Studies. ACS OMEGA 2022; 7:3775-3795. [PMID: 35128286 PMCID: PMC8811919 DOI: 10.1021/acsomega.1c06694] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/11/2022] [Indexed: 05/04/2023]
Abstract
In the present work, a concise library of 1,3,5-triaryl-2-pyrazolines (2a-2q) was designed and synthesized by employing a multistep strategy, and the newly synthesized compounds were screened for their urease and α-glucosidase inhibitory activities. The compounds (2a-2q) were characterized using a combination of several spectroscopic techniques including FT-IR, 1H NMR, 13C NMR, and EI-MS. All the synthesized compounds, except compound 2i, were potent against urease as compared to the standard inhibitor thiourea (IC50 = 21.37 ± 0.26 μM). These analogs disclosed varying degrees of urease inhibitory activities ranging from 9.13 ± 0.25 to 18.42 ± 0.42 μM. Compounds 2b, 2g, 2m, and 2q having IC50 values of 9.36 ± 0.27, 9.13 ± 0.25, 9.18 ± 0.35, and 9.35 ± 0.35 μM, respectively, showed excellent inhibitory activity as compared to standard thiourea (IC50 = 21.37 ± 0.26 μM). A kinetic study of compound 2g revealed that compound 2g inhibited urease in a competitive mode. Among the synthesized pyrazolines, the compounds 2c, 2k, 2m, and 2o exhibited excellent α-glucosidase inhibitory activity with the lowest IC50 values of 212.52 ± 1.31, 237.26 ± 1.28, 138.35 ± 1.32, and 114.57 ± 1.35 μM, respectively, as compared to the standard acarbose (IC50 = 375.82 ± 1.76 μM). The compounds (2a-2q) showed α-glucosidase IC50 values in the range of 114.57 ± 1.35 to 462.94 ± 1.23 μM. Structure-activity relationship revealed that the size and electron-donating or -withdrawing effects of substituents influenced the activities, which led to the urease and α-glucosidase inhibiting properties. Compound 2m was a dual potent inhibitor against urease and α-glucosidase due to the presence of 2-CF3 electron-withdrawing functionality on the phenyl ring. To the best of our knowledge, these synthetic compounds were found to be the most potent dual inhibitors of urease and α-glucosidase with minimum IC50 values. The cytotoxicity of the compounds (2a-2q) was also investigated against human cell lines MCF-7 and HeLa. Compound 2l showed moderate cytotoxic activity against MCF-7 and HeLa cell lines. Moreover, in silico studies on most active compounds were also performed to understand the binding interaction of most active compounds with active sites of urease and α-glucosidase enzymes. Some compounds exhibited drug-like characteristics due to their lower cytotoxic and good ADME profiles.
Collapse
Affiliation(s)
- Rabia Mehmood
- Department
of Chemistry, Govt. College Women University, Sialkot 51300, Pakistan
| | - Amina Sadiq
- Department
of Chemistry, Govt. College Women University, Sialkot 51300, Pakistan
| | - Reem I. Alsantali
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | | | - Meshari A. Alsharif
- Department
of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Nafeesa Naeem
- Department
of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | - Asif Javid
- Department
of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | - Munirah M. Al-Rooqi
- Department
of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Gul-e-Saba Chaudhry
- Institute
of Marine Biotechnology, Universiti Malaysia
Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
- Microbiology
and Biotechnology Research Lab, Fatima Jinnah
Women University, Rawalpindi 23451, Pakistan
| | - Saleh A. Ahmed
- Department
of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
- Department
of Chemistry, Faculty of Science, Assiut
University, Assiut 71516, Egypt
| |
Collapse
|