1
|
Ayan EK, Çoban G, Soyer Z. Design, synthesis, biological evaluation, and molecular modeling studies of some quinazolin-4(3 H)-one-benzenesulfonamide hybrids as potential α-glucosidase inhibitors. J Biomol Struct Dyn 2024:1-21. [PMID: 39539169 DOI: 10.1080/07391102.2024.2427373] [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: 01/25/2024] [Accepted: 07/30/2024] [Indexed: 11/16/2024]
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by hyperglycemia, posing serious health risks and becoming increasingly prevalent. Prolonged hyperglycemia can lead to complications such as nephropathy, neuropathy, retinopathy, cardiovascular damage, and blindness. Controlling hyperglycemia through α-glucosidase inhibitors, which slow down carbohydrate breakdown, is an effective treatment strategy. However, current inhibitors like acarbose, voglibose, and miglitol while used to manage type 2 diabetes, have significant side effects. Therefore, developing new α-glucosidase inhibitors that are more effective and have fewer side effects is crucial. In this study, a series of novel quinazolin-4(3H)-one-benzenesulfonamide hybrid compounds were designed, synthesized, and evaluated for in vitro α-glucosidase inhibitory activity. The compounds showed higher enzyme inhibition potency, with IC50 values ranging between 129.2 ± 0.5 and 558.7 ± 13.7 µM, compared to acarbose (IC50=814.3 ± 13.5 µM). Among the tested compounds, compound 10, bearing a 4-chlorophenyl ring on the nitrogen atom of the sulfonamide group, was the most active, with an IC50 value of 129.2 ± 0.5 µM. Enzyme kinetics analyses and molecular modeling studies were conducted to understand their inhibition mechanisms and interactions with the enzyme. The kinetic studies revealed a mixed-type inhibition model, indicating that the compounds bind to the enzyme-substrate complex with higher affinity than to the free enzyme. Molecular modeling results confirmed these findings. Additionally, in silico prediction studies showed that the selected compounds have favourable physicochemical and drug-like properties. These results suggest these compounds have potential for further optimization and development as effective α-glucosidase inhibitors for diabetes treatment.
Collapse
Affiliation(s)
- Emre Kadir Ayan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Süleyman Demirel University, Isparta, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey
| | - Güneş Çoban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey
| | - Zeynep Soyer
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey
| |
Collapse
|
2
|
Peytam F, Hosseini FS, Fathimolladehi R, Nayeri MJD, Moghadam MS, Bayati B, Norouzbahari M, Foroumadi R, Bonyasi F, Divsalar R, Mojtabavi S, Faramarzi MA, Tehrani MB, Firoozpour L, Foroumadi A. Design, synthesis, and evaluation of novel substituted imidazo[1,2-c]quinazoline derivatives as potential α-glucosidase inhibitors with bioactivity and molecular docking insights. Sci Rep 2024; 14:27507. [PMID: 39528585 PMCID: PMC11555253 DOI: 10.1038/s41598-024-78878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
α-Glucosidase inhibitors are important in the treatment of type 2 diabetes by regulating blood glucose levels and reducing carbohydrate absorption. The present study focuses on identifying new inhibitors bearing imidazo[1,2-c]quinazoline backbone through multi-step synthesis. The inhibitory potencies of the novel derivatives were tested against Saccharomyces cerevisiae α-glucosidase, revealing IC50 values ranging from 50.0 ± 0.12 µM to 268.25 ± 0.09 µM. Among them, 2-(4-(((2,3-diphenylimidazo[1,2-c]quinazolin-5-yl)thio)methyl)-1H-1,2,3-triazol-1-yl)-N-(2-methoxyphenyl)acetamide (19e) and 2-(4-((benzo[4,5]imidazo[1,2-c]quinazolin-6-ylthio)methyl)-1H-1,2,3-triazol-1-yl)-N-(2-methoxyphenyl)acetamide (27e) emerged as the most potent inhibitors and were further investigated in various assessments. Finally, molecular docking studies were performed to reveal the crucial binding interactions and to confirm the results obtained from structure-activity relationship (SAR) analysis.
Collapse
Affiliation(s)
- Fariba Peytam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Sadat Hosseini
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Fathimolladehi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahdis Sadeghi Moghadam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Bayati
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Norouzbahari
- Faculty of Pharmacy, Final International University, Catalkoy, Kyrenia via Mersin 10 Turkey, Turkish Republic of Northern Cyprus
| | - Roham Foroumadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Bonyasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ruzbehan Divsalar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Barazandeh Tehrani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
3
|
Kushavah U, Mahapatra PP, Ahmed S, Siddiqi MI. Pharmacophore-based 3D-QSAR modeling, virtual screening, docking, molecular dynamics and biological evaluation studies for identification of potential inhibitors of alpha-glucosidase. J Mol Model 2024; 30:389. [PMID: 39476191 DOI: 10.1007/s00894-024-06181-y] [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: 05/23/2023] [Accepted: 10/14/2024] [Indexed: 11/14/2024]
Abstract
CONTEXT Alpha-glucosidase enzyme is considered an important therapeutic target for controlling hyperglycemia associated with type 2 diabetes. Novel scaffolds identified as potential alpha-glucosidase inhibitors from the Maybridge library utilizing pharmacophore modeling, molecular docking and biological evaluation are reported in this manuscript. METHOD A total of 51 xanthone series scaffolds previously reported as alpha-glucosidase inhibitors were collected and used as training and test sets. These sets were employed to develop and validate a pharmacophore-based 3D-QSAR model with statistically meaningful results using Schrodinger software. The model showed a high F value (F, 80.1) at five component partial least square factors, a high cross-validation coefficient (Q2, 0.66) and a good correlation coefficient (R2, 0.95). Pearson correlation coefficient (r) of 0.8400 indicated a greater degree of confidence in the model. Subsequently, virtual screening was performed with PHASE module of Schrodinger software using the above model to identify novel alpha-glucosidase inhibitors, and mapped compounds were evaluated for their interactions with the protein. The X-ray co-crystallised structure of the alpha-glucosidase protein in complex with acarbose (PDB Code: 5NN8) was used for molecular docking analysis using GLIDE module and a total of eight compounds were further selected for biological evaluation. Molecular dynamics analysis using GROMACS software was performed in the active site of alpha-glucosidase protein to gain insights into binding mechanism of the four active compounds which were finally found to exhibit inhibitory activity in the biological assay.
Collapse
Affiliation(s)
- Unnati Kushavah
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pinaki Prasad Mahapatra
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shakil Ahmed
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohammad Imran Siddiqi
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
4
|
Hameed S, Saleem F, Özil M, Baltaş N, Salar U, Ashraf S, Ul-Haq Z, Taha M, Khan KM. Indenoquinoxaline-phenylacrylohydrazide hybrids as promising drug candidates for the treatment of type 2 diabetes: In vitro and in silico evaluation of enzyme inhibition and antioxidant activity. Int J Biol Macromol 2024; 263:129517. [PMID: 38266833 DOI: 10.1016/j.ijbiomac.2024.129517] [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: 07/27/2023] [Revised: 12/26/2023] [Accepted: 01/13/2024] [Indexed: 01/26/2024]
Abstract
Existing drugs that are being used to treat type-2 diabetes mellitus are associated with several side effects; thus, exploring potential drug candidates is still an utter need these days. Hybrids of indenoquinoxaline and hydrazide have never been explored as antidiabetic agents. In this study, a series of new indenoquinoxaline-phenylacrylohydrazide hybrids (1-30) were synthesized, structurally characterized, and evaluated for α-amylase and α-glucosidase inhibitory activities, as well as for their antioxidant properties. All scaffolds exhibited varying degrees of inhibitory activity against both enzymes, with IC50 values ranging from 2.34 to 61.12 μM for α-amylase and 0.42 to 54.72 μM for α-glucosidase. Particularly, compounds 10, 16, 17, 18, 24, and 25 demonstrated the highest efficacy in inhibiting α-amylase, while compounds 6, 7, 8, 10, 12, 14, 13, 16, 17, 18, 24, and 25 were the most effective α-glucosidase inhibitors, compared to standard acarbose. Moreover, most of these compounds displayed substantial antioxidant potential compared to standard butylated hydroxytoluene (BHT). Kinetics studies revealed competitive inhibition modes by compounds. Furthermore, a comprehensive in silico study and toxicity prediction were also conducted, further validating these analogs as potential drug candidates. The structured compounds demonstrated enhanced profiles, underscoring their potential as primary candidates in drug discovery.
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
| | - Faiza Saleem
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Musa Özil
- Department of Chemistry, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Nimet Baltaş
- Department of Chemistry, Recep Tayyip Erdogan University, 53100 Rize, 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
| | - Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - 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
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Pakistan Academy of Science, 3-Constitution Avenue, G-5/2, Islamabad 44000, Pakistan.
| |
Collapse
|
5
|
Ullah S, Halim SA, Waqas M, Mansoor F, Avula SK, Khan FA, Perviaz M, Ogaly HA, Khan A, Al-Harrasi A. Biochemical and computational inhibition of α-glucosidase by novel metronidazole-linked 1 H-1,2,3-triazole and carboxylate moieties: kinetics and dynamic investigations. J Biomol Struct Dyn 2024:1-21. [PMID: 38433423 DOI: 10.1080/07391102.2024.2322622] [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: 09/19/2023] [Accepted: 02/16/2024] [Indexed: 03/05/2024]
Abstract
In the current study, metronidazole derivatives containing 1H-1,2,3-triazole and carboxylate moieties were evaluated in vitro and by computational methods for their anti-diabetic potential to insight into their medicinal use for the management of type II diabetes mellitus. Interestingly all 14 compounds displayed high to significant inhibitory capability against the key carbohydrate's digestive enzyme α-glucosidase with IC50 values in range of 9.73-56.39 μM, as compared to marketed drug acarbose (IC50 = 873.34 ± 1.67 μM). Compounds 5i and 7c exhibited the highest inhibition, therefore, these two compounds were further evaluated for their mechanistic studies to explore its type of inhibition. Compounds 5i and 7c both displayed a concentration-dependent (competitive type of inhibition) with Ki values 7.14 ± 0.01, 6.15 ± 0.02 μM, respectively, which conclude their favourable interactions with the active site residues of the α-glucosidase. Interestingly all compounds are non-cytotoxic against BJ cell line. To further validate our findings, in-silico approaches like molecular docking, and molecular dynamic simulations were applied to investigate the mode of bindings of compounds with the enzyme and identifies their inhibition mechanism, which strongly complements our experimental findings.
Collapse
Affiliation(s)
- Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Farheen Mansoor
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Satya Kumar Avula
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Farhan A Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, KPK, Pakistan
| | - Muhammad Perviaz
- Department of Basic & Applied Chemistry, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Hanan A Ogaly
- Chemistry Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
6
|
Zhang Y, Li Y, Zhai Y, Zhao X, Lv M, Yu S, Xiao H, Song Y. Inhibitory mechanism of chrysin and diosmetin to α-glucosidase: insights from kinetics, multispectroscopy and molecular docking investigations. J Biomol Struct Dyn 2024:1-13. [PMID: 38289727 DOI: 10.1080/07391102.2024.2310207] [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: 09/01/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
Inhibition of α-glucosidase activity is a promising method to prevent postprandial hyperglycemia. The inhibitory effect and interaction of chrysin and diosmetin on α-glucosidase were studied in this study. The results of inhibition kinetics showed that chrysin and diosmetin reversibly inhibited α-glucosidase activity with IC50 value of 26.445 ± 1.406 μmol L-1 and 18.380 ± 1.264 μmol L-1, respectively. Further research revealed that chrysin exhibited a mixed-type inhibitory pattern against α-glucosidase, while diosmetin was noncompetitive inhibitory with Ki value of (2.6 ± 0.04) ×10-4 mol L-1. Fluorescence spectroscopy showed that both chrysin and diosmetin could quench the intrinsic fluorescence of α-glucosidase, the maximum emission wavelength of tyrosine (Tyr) and tryptophan (Trp) were not moved by chrysin, but red shifted by diosmetin. UV-Vis, fourier transform infrared spectroscopy (FT-IR) and circular dichroism (CD) measurements showed that the secondary structure and microenvironment of α-glucosidase were changed by chrysin and diosmetin. Further analysis of molecular docking showed that chrysin and diosmetin could bind with α-glucosidase and might cause the decrease of α-glucosidase activity. The results of molecular dynamics (MD) simulation showed that the stability of chrysin (or diosmetin)-α-glucosidase complex system was changed during binding process. In conclusion, chrysin and diosmetin are good α-glucosidase inhibitors.
Collapse
Affiliation(s)
- Yuqing Zhang
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Yaping Li
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Yuhan Zhai
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Xing Zhao
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Mingxing Lv
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Shaoxuan Yu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Haifang Xiao
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| | - Yuanda Song
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong, China
| |
Collapse
|
7
|
Peytam F, Hosseini FS, Hekmati M, Bayati B, Moghadam MS, Emamgholipour Z, Firoozpour L, Mojtabavi S, Faramarzi MA, Sadat-Ebrahimi SE, Tehrani MB, Foroumadi A. Imidazo[1,2-c]quinazolines as a novel and potent scaffold of α-glucosidase inhibitors: design, synthesis, biological evaluations, and in silico studies. Sci Rep 2023; 13:15672. [PMID: 37735489 PMCID: PMC10514295 DOI: 10.1038/s41598-023-42549-5] [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: 05/29/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
α-Glucosidase inhibition is an approved treatment for type 2 diabetes mellitus (T2DM). In an attempt to develop novel anti-α-glucosidase agents, two series of substituted imidazo[1,2-c]quinazolines, namely 6a-c and 11a-o, were synthesized using a simple, straightforward synthetic routes. These compounds were thoroughly characterized by IR, 1H and 13C NMR spectroscopy, as well as mass spectrometry and elemental analysis. Subsequently, the inhibitory activities of these compounds were evaluated against Saccharomyces cerevisiae α-glucosidase. In present study, acarbose was utilized as a positive control. These imidazoquinazolines exhibited excellent to great inhibitory potencies with IC50 values ranging from 12.44 ± 0.38 μM to 308.33 ± 0.06 μM, which were several times more potent than standard drug with IC50 value of 750.0 ± 1.5 μM. Representatively, compound 11j showed remarkable anti-α-glucosidase potency with IC50 = 12.44 ± 0.38 μM, which was 60.3 times more potent than positive control acarbose. To explore the potential inhibition mechanism, further evaluations including kinetic analysis, circular dichroism, fluorescence spectroscopy, and thermodynamic profile were carried out for the most potent compound 11j. Moreover, molecular docking studies and in silico ADME prediction for all imidazoquinazolines 6a-c and 11a-o were performed to reveal their important binding interactions, as well as their physicochemical and drug-likeness properties, respectively.
Collapse
Affiliation(s)
- Fariba Peytam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Sadat Hosseini
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Malak Hekmati
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Bahareh Bayati
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdis Sadeghi Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Maliheh Barazandeh Tehrani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Dutta N, Dutta B, Dutta A, Sarma B, Sarma D. Room temperature ligand-free Cu 2O-H 2O 2 catalyzed tandem oxidative synthesis of quinazoline-4(3 H)-one and quinazoline derivatives. Org Biomol Chem 2023; 21:748-753. [PMID: 36602007 DOI: 10.1039/d2ob02085h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
An efficient and simple copper catalytic system has been developed for the synthesis of medicinally important 2-substituted quinazoline-4(3H)-ones from 2-aminobenzonitrile and benzyl alcohol derivatives and additionally 2-substituted quinazolines from 2-aminobenzylamine and benzaldehyde derivatives. Mild oxidant H2O2 was utilized, providing excellent product yields. The molecular structure of one of the compounds was substantiated through SC-XRD. The versatility of the protocol was demonstrated through gram-scale syntheses.
Collapse
Affiliation(s)
- Nilakshi Dutta
- Department of Chemistry, Dibrugarh University, Dibrugarh-786004, Assam, India.
| | - Bidyutjyoti Dutta
- Department of Chemistry, Dibrugarh University, Dibrugarh-786004, Assam, India.
| | - Apurba Dutta
- Department of Chemistry, DHSK College, Dibrugarh-786001, Assam, India
| | - Bipul Sarma
- Department of Chemical Sciences, Tezpur University, Tezpur-784028, Assam, India
| | - Diganta Sarma
- Department of Chemistry, Dibrugarh University, Dibrugarh-786004, Assam, India.
| |
Collapse
|
9
|
Babatunde O, Hameed S, Mbachu K, Saleem F, Chigurupati S, Wadood A, Ur R, Venugopal V, Khan K, Taha M, Ekundayo O, Khan M. Evaluation of derivatives of 2,3-dihydroquinazolin-4(1H)-one as inhibitors of cholinesterases and their antioxidant activity: In vitro, in silico, and kinetics studies. JOURNAL OF THE SERBIAN CHEMICAL SOCIETY 2023; 88:825-840. [DOI: 10.2298/jsc211106005b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
In search of potent inhibitors of cholinesterase enzymes and antioxidant agents, synthetic derivatives of dihydroquinazolin-4(1H)-one (1?38) were evaluated as potential anti-Alzheimer agents through in vitro acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitions and radical (DPPH and ABTS) scavenging activities. The structure?activity relationship (SAR) was mainly based on the different substituents at the aryl part which showed a significant effect on the inhibitory potential of enzymes and radical scavenging activities. The kinetic studies of most active compounds showed a noncompetitive mode of inhibition for AChE and a competitive mode of inhibition for the BChE enzyme. Additionally, molecular modelling studies were carried out to investigate the possible binding interactions of quinazolinone derivatives 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, Pakistan
| | - Shehryar Hameed
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Kingsley Mbachu
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan + Department of Chemistry, University of Ibadan, Nigeria
| | - Faiza Saleem
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, UCSS, Abdul Wali Khan University, Mardan, Pakistan
| | - Rehman Ur
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, UCSS, Abdul Wali Khan University, Mardan, Pakistan
| | | | - Khalid Khan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan + Department of Clinical Pharmacy, 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
| | | | - Maria Khan
- Third World Center for Science and Technology, H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
10
|
Hameed S, Babatunde O, Salar U, Jabbar A, Chigurupati S, Solangi M, Atta L, Ul-Haq Z, Saleem F, Bhatia S, Al-Harrasi A, Taha M, Khan KM. 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: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/15/2022] [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
Affiliation(s)
- Shehryar Hameed
- H. E. J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Oluwatoyin Babatunde
- H. E. J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Uzma Salar
- Dr. Panjwani Center for Molecular Medicine & Drug Research, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary & Agricultural Sciences, The University of Melbourne, 250 Princes Highway, Werribee, Victoria, 3030, Australia
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Mehwish Solangi
- H. E. J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Lubna Atta
- Dr. Panjwani Center for Molecular Medicine & Drug Research, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine & Drug Research, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Faiza Saleem
- H. E. J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mauz, Nizwa, P.O. Box 33, Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mauz, Nizwa, P.O. Box 33, Oman
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, P.O. Box 1982, 31441, Saudi Arabia
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Department of Clinical Pharmacy, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, P.O. Box 1982, 31441, Saudi Arabia
| |
Collapse
|
11
|
Kulsoom U, Salar U, Khan KM, Chigurupati S, Syed S, Wadood A, Rehman AU, Fatima B, Saleem F, Taha M, Felemban SG, Dachani SR, Perveen S. Synthesis and evaluation of aryl aminomethylene substituted barbiturates and thiobarbiturates as novel α-amylase inhibitors and radical scavengers. MONATSHEFTE FÜR CHEMIE - CHEMICAL MONTHLY 2022; 153:949-960. [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] [Received: 03/07/2022] [Accepted: 08/09/2022] [Indexed: 11/27/2022]
|
12
|
Newly Designed Quinazolinone Derivatives as Novel Tyrosinase Inhibitor: Synthesis, Inhibitory Activity, and Mechanism. Molecules 2022; 27:molecules27175558. [PMID: 36080324 PMCID: PMC9457556 DOI: 10.3390/molecules27175558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022] Open
Abstract
We synthesized a series of quinazolinone derivates as tyrosinase inhibitors and evaluated their inhibition constants. We synthesized 2-(2,6-dimethylhepta-1,5-dien-1-yl)quinazolin-4(3H)-one (Q1) from the natural citral. The concentration, which led to 50% activity loss of Q1, was 103 ± 2 μM (IC50 = 103 ± 2 μM). Furthermore, we considered Q1 to be a mixed-type and reversible tyrosinase inhibitor, and determined the KI and KIS inhibition constants to be 117.07 μM and 423.63 μM, respectively. Our fluorescence experiment revealed that Q1 could interact with the substrates of tyrosine and L-DOPA in addition to tyrosinase. Molecular docking studies showed that the binding of Q1 to tyrosinase was driven by hydrogen bonding and hydrophobicity. Briefly, the current study confirmed a new tyrosinase inhibitor, which is expected to be developed into a novel pigmentation drug.
Collapse
|
13
|
Karrouchi K, Sert Y, Ansar M, Radi S, El Bali B, Imad R, Alam A, Irshad R, Wajid S, Altaf M. Synthesis, α-Glucosidase Inhibition, Anticancer, DFT and Molecular Docking Investigations of Pyrazole Hydrazone Derivatives. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2097275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Khalid Karrouchi
- Faculty of Medicine and Pharmacy, Laboratory of Analytical Chemistry and Bromatology, Mohammed V University in Rabat, Rabat, Morocco
| | - Yusuf Sert
- Science and Art Faculty, Department of Physics, Sorgun Vocational School, Yozgat Bozok University, Yozgat, Turkey
| | - M’hammed Ansar
- Laboratory of Medicinal Chemistry, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Morocco
| | - Smaail Radi
- Laboratory of Applied Chemistry and Environment (LCAE), Faculty of Sciences, Department of Chemistry, University Mohammed Premier, Oujda, Morocco
| | - Brahim El Bali
- Laboratory of Organic, Macromolecular Chemistry and Natural Products, Faculty of Sciences, Mohammed I University, Oujda, Morocco
| | - Rehan Imad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Anum Alam
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rimsha Irshad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Sheeba Wajid
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biology Science, University of Karachi, Karachi, Pakistan
| | - Muhammad Altaf
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biology Science, University of Karachi, Karachi, Pakistan
| |
Collapse
|
14
|
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: 17] [Impact Index Per Article: 5.7] [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
|
15
|
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 2022; 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] [Grants] [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
|
16
|
Satyanarayana N, Boddu R, Sathish K, Nagaraju S, K D, Pawar R, Shirisha T, Kashinath D. Synthesis of 2-styryl-quinazoline and 3-styryl-quinoxaline based sulfonate esters via sp3 C-H activation and their evaluation for α-glucosidase inhibition. NEW J CHEM 2022. [DOI: 10.1039/d1nj05644a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synthesis of 2-styryl-quinazolines and 3-styryl-quinoxaline based sulfonates is reported via sp3 C-H functionalization in the presence of triethylamine (10 mol%). The resulting compounds were tested for the α-glucosidase enzyme inhibition...
Collapse
|
17
|
New biologically dynamic hybrid pharmacophore triazinoindole-based-thiadiazole as potent α-glucosidase inhibitors: In vitro and in silico study. Int J Biol Macromol 2021; 199:77-85. [PMID: 34968547 DOI: 10.1016/j.ijbiomac.2021.12.147] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 12/02/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
Triazinoindole bearing thiadiazole derivatives (1-25) have been synthesized and characterized through different spectroscopic techniques such as 1H, 13C-NMR and HREI-MS. The purpose of the study was to investigate the anti-diabetic activity of the synthesized triazinoindole bearing thiadiazole derivatives by inhibition of α-glucosidase. All synthesized analogues showed outstanding inhibition of α-glucosidase enzyme with IC50 values ranging from 2.5 ± 0.10 to 38.10 ± 0.10 µM as compared to the standard drug acarbose (IC50 = 38.45 ± 0.80 µM). Analogue 4 (IC50 = 2.5 ± 0.10 µM) was identifies as the most potent analogue in the series with fifteen folds more active than standard acarbose. Structure activity relationship (SAR) studies suggested that α-glucosidase activities of triazinoindole bearing thiadiazole are primarily dependent upon on number and position of different substitutions present on phenyl parts. Molecular docking study were conducted of the optimized compounds (i.e., compound 4, 6, and 3 etc. using MOE default parameters), the results revealed that compound 4, 6, and 3 showed numerous key interactions with the target protein, which indicate the high potential of these compounds against the target compound. All these compounds were screened for cytotoxic activity against normal normal Vero cell line and found non-toxic.
Collapse
|
18
|
Azimi F, Azizian H, Najafi M, Hassanzadeh F, Sadeghi-Aliabadi H, Ghasemi JB, Ali Faramarzi M, Mojtabavi S, Larijani B, Saghaei L, Mahdavi M. Design and synthesis of novel quinazolinone-pyrazole derivatives as potential α-glucosidase inhibitors: Structure-activity relationship, molecular modeling and kinetic study. Bioorg Chem 2021; 114:105127. [PMID: 34246971 DOI: 10.1016/j.bioorg.2021.105127] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/11/2023]
Abstract
In this study, a new series of quinazolinone-pyrazole hybrids were designed, synthesized and screened for their α-glucosidase inhibitory activity. The results of the in vitro screening indicated that all the molecular hybrids exhibited more inhibitory activity (IC50 values ranging from 60.5 ± 0.3 µM-186.6 ± 20 μM) in comparison to standard acarbose (IC50 = 750.0 ± 10.0 µM). Limited structure-activity relationship suggested that the variation in the inhibitory activities of the compounds affected by different substitutions on phenyl rings of diphenyl pyrazole moiety. The enzyme kinetic studies of the most potent compound 9i revealed that it inhibited α-glucosidase in a competitive mode with a Ki of 56 μM. Molecular docking study was performed to predict the putative binding interaction. As expected, all pharmacophoric moieties used in the initial structure design playing a pivotal role in the interaction with the binding site of the enzyme. In addition, by performing molecular dynamic investigation and MM-GBSA calculation, we investigated the difference in structural perturbation and dynamic behavior that is observed over α-glycosidase in complex with the most active compound and acarbose relative to unbound α-glycosidase enzyme.
Collapse
Affiliation(s)
- Fateme Azimi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461 Isfahan, Iran
| | - Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Science, Tehran, Iran
| | - Mohammad Najafi
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461 Isfahan, Iran
| | - Hojjat Sadeghi-Aliabadi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461 Isfahan, Iran
| | - Jahan B Ghasemi
- School of Chemistry, University College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran
| | - Mohammad Ali Faramarzi
- 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
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Lotfollah Saghaei
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461 Isfahan, Iran.
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
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
|
20
|
Shaheen A, Ashiq U, Jamal RA, Khan KM, Gul S, Yousuf S, Ali ST. Design and Synthesis of Fluoroquinolone Derivatives as Potent α‐Glucosidase Inhibitors: In Vitro Inhibitory Screening with In Silico Docking Studies. ChemistrySelect 2021. [DOI: 10.1002/slct.202003956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Aasia Shaheen
- Department of Chemistry University of Karachi Karachi 75270 Pakistan
| | - Uzma Ashiq
- Department of Chemistry University of Karachi Karachi 75270 Pakistan
| | - Rifat Ara Jamal
- Department of Chemistry 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 1982 Dammam 31441 Saudi Arabia
| | - Sana Gul
- Department of Chemistry Federal Urdu University of Art, Science and Technology Karachi Pakistan
| | - Sammer Yousuf
- H. E. J. Research Institute of Chemistry International Center for Chemical and Biological Sciences University of Karachi Karachi 75270 Pakistan
| | - Syed Tahir Ali
- Department of Chemistry Federal Urdu University of Art, Science and Technology Karachi Pakistan
| |
Collapse
|
21
|
Sherafati M, Mirzazadeh R, Barzegari E, Mohammadi-Khanaposhtani M, Azizian H, Sadegh Asgari M, Hosseini S, Zabihi E, Mojtabavi S, Ali Faramarzi M, Mahdavi M, Larijani B, Rastegar H, Hamedifar H, Hamed Hajimiri M. Quinazolinone-dihydropyrano[3,2-b]pyran hybrids as new α-glucosidase inhibitors: Design, synthesis, enzymatic inhibition, docking study and prediction of pharmacokinetic. Bioorg Chem 2021; 109:104703. [PMID: 33609917 DOI: 10.1016/j.bioorg.2021.104703] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/29/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023]
Abstract
A series of new quinazolinone-dihydropyrano[3,2-b]pyran derivatives 10A-L were synthesized by simple chemical reactions and were investigated for inhibitory activities against α-glucosidase and α-amylase. New synthesized compounds showed high α-glucosidase inhibition effects in comparison to the standard drug acarbose and were inactive against α-amylase. Among them, the most potent compound was compound 10L (IC50 value = 40.1 ± 0.6 µM) with inhibitory activity around 18.75-fold more than acarboase (IC50 value = 750.0 ± 12.5 µM). This compound was a competitive inhibitor into α-glucosidase. Our obtained experimental results were confirmed by docking studies. Furthermore, the cytotoxicity of the most potent compounds 10L, 10G, and 10N against normal fibroblast cells and in silico druglikeness, ADME, and toxicity prediction of these compounds were also evaluated.
Collapse
Affiliation(s)
- Maedeh Sherafati
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Barzegari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | | | - Samanesadat Hosseini
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 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
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - 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
| | - Mir Hamed Hajimiri
- Nano Alvand Company, Avicenna Tech Park, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Synthesis, in vitro, and in silico studies of newly functionalized quinazolinone analogs for the identification of potent α-glucosidase inhibitors. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2021. [DOI: 10.1007/s13738-021-02159-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
23
|
Kausar N, Ullah S, Khan MA, Zafar H, Atia-Tul-Wahab, Choudhary MI, Yousuf S. Celebrex derivatives: Synthesis, α-glucosidase inhibition, crystal structures and molecular docking studies. Bioorg Chem 2020; 106:104499. [PMID: 33288319 DOI: 10.1016/j.bioorg.2020.104499] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
Celebrex (1), commonly used as an anti-inflammatory drug, was functionalized (compounds 2-9) to identify new α-glucosidase inhibitors. Initially, all the synthesized derivatives were evaluated for anti-inflammatory activity but none was found to be active. Subsequently a random biological screening was carried out. Interestingly many of them were found to be potent α-glucosidase inhibitors in vitro. All the structures of synthesized derivatives were deduced through 1H NMR, FAB-MS, HR-MS, FT-IR analysis. The single-crystal X-ray structures of compounds 1, and 5 further confirmed the assigned structures. Compounds exhibited a potent α-glucosidase inhibitory activity (IC50 = 92.32 ± 1.530-445.20 ± 1.04 µM) against tested standard acarbose (IC50 = 875.75 ± 2.08 µM), except compounds 2 and 4, which appeared as inactive. Among them, compound 9 (IC50 = 92.32 ± 1.530 µM) was the most potent inhibitor of α-glucosidase enzyme. Molecular docking studies revealed that compounds 6, and 9 interacted with the key amino acid residues of α-glucosidase via H-bonding, and π-π stacking interactions. α-Glucosidase is a key target for the anti-diabetic drug development, and its inhibitors are known to exert anti hyperglycemic effect and help in lowering of post-prandial blood glucose levels.
Collapse
Affiliation(s)
- Nabeela Kausar
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Saeed Ullah
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Maria Aqeel Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Humaira Zafar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Atia-Tul-Wahab
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - M Iqbal Choudhary
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21412, Saudi Arabia; Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Komplek Campus C, Surabaya 60115, Indonesia
| | - Sammer Yousuf
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
24
|
Liu D, Cao X, Kong Y, Mu T, Liu J. Inhibitory mechanism of sinensetin on α-glucosidase and non-enzymatic glycation: Insights from spectroscopy and molecular docking analyses. Int J Biol Macromol 2020; 166:259-267. [PMID: 33115652 DOI: 10.1016/j.ijbiomac.2020.10.174] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/03/2020] [Accepted: 10/21/2020] [Indexed: 01/06/2023]
Abstract
Inhibition of α-glucosidase and non-enzymatic glycation is regarded as an effective method to prevent and treat type 2 diabetes and its complications. In this study, the inhibition of sinensetin on α-glucosidase and non-enzymatic glycation was studied with multi-spectroscopic techniques and molecular docking analysis. The results of fluorescence spectroscopy analysis indicated that sinensetin quenched the endogenous fluorescence of α-glucosidase in static manner. The binding of sinensetin with α-glucosidase was a spontaneous process primarily driven by hydrophobic interaction. At 298 K, the binding constant was (5.70 ± 0.12) × 104 L·mol-1 and the binding site number was 1. The conformation of α-glucosidase was altered by sinensetin, which was revealed by circular dichroism (CD), FTIR spectra, synchronous fluorescence and three-dimensional (3D) fluorescence spectroscopy methods. Molecular docking analysis demonstrated that sinensetin interacted with the amino acid residues of α-glucosidase, which might prevent the entrance of substrate, leading to the decrease of catalytic efficiency of α-glucosidase. Furthermore, glycation assays showed that sinensetin stabilized the structure of bovine serum albumins (BSA), interacted with BSA, strongly inhibited the formation of dityrosine, N'-formylkynurenine and advanced glycation end products (AGEs). This study provided useful information concerning sinensetin preventing and treating type 2 diabetes and its related complications.
Collapse
Affiliation(s)
- Dan Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, PR China
| | - Xiangyu Cao
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, PR China
| | - Yuchi Kong
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, PR China
| | - Teng Mu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, PR China
| | - Jianli Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, PR China.
| |
Collapse
|
25
|
Bule MH, Esfandyari R, Tafesse TB, Amini M, Faramarzi MA, Abdollahi M. Synthesis, Molecular Docking and α-Glucosidase Inhibitory Activity Study of 2,4,6-triaryl Pyrimidine Derivatives. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817666200103130536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
α-Glucosidase inhibitors hinder the carbohydrate digestion and play an
important role in the treatment of diabetes mellitus. α-glucosidase inhibitors available on the market
are acarbose, miglitol, and voglibose. However, the use of acarbose is diminishing due to related
side effects like diarrhea, bloating and abdominal distension.
Objectives:
This study aimed to synthesize 2,4,6-triaryl pyrimidines derivatives, screen their α-
glucosidase inhibitory activity, perform kinetic and molecular docking studies.
Methods:
A series of 2,4,6-triaryl pyrimidine derivatives were synthesized and their α-glucosidase
inhibitory activity was screened in vitro. Pyrimidine derivatives 4a-m were synthesized via a twostep
reaction with a yield between 49 and 93%. The structure of the synthesized compounds was
confirmed by different spectroscopic techniques (IR, NMR and MS). The in vitro α-glucosidase
inhibition activities of the synthesized compounds 4a-m was also evaluated against Saccharomyces
cerevisiae α-glucosidase.
Results and Discussion:
The majority of synthesized compounds had α-glucosidase inhibitory
activity. Particularly compounds 4b and 4g were the most active compounds with an IC50 value of
125.2± 7.2 and 139.8 ± 8.1 μM respectively. The kinetic study performed for the most active
compound 4b revealed that the compound was a competitive inhibitor of Saccharomyces cerevisiae
α-glucosidase with Ki of 122 μM. The molecular docking study also revealed that the two
compounds have important binding interactions with the enzyme active site.
Conclusion:
2,4,6-triarylpyrimidine derivative 4a-m were synthesized and screened for α-
glucosidase inhibitory activity. Most of the synthesized compounds possess α-glucosidase inhibitory
activity, and compound 4b demonstrated the most significant inhibitory action as compared to
acarbose.
Collapse
Affiliation(s)
- Mohammed Hussen Bule
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Roghaieh Esfandyari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tadesse Bekele Tafesse
- Department of Medicinal Chemistry, 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 Ali Faramarzi
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
jain N, Singour PK. Novel 3-Substituted-2, 3-Dihydro-2-Thioxoquinazolin-4-(1H)-one derivative as Anticonvulsants: Synthesis, Molecular Docking and Pharmacological Screening. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666191024090857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
According to the World Health Organization, 50 million people worldwide
are suffering from epilepsy, making it one of the most common neurological diseases globally. 2,3
disubstituted quinazolinone-4-one derivatives endowed with various pharmacological activity, particularly
having anticonvulsant action.
Objectives:
The aim of this study was to synthesize 3-Substituted-2,3-Dihydro-2-thioxoquinazolin-
4-(1H)-one derivative and evaluate for anticonvulsant activity and neurotoxicity in order to find an
efficient, compound with lesser side effects.
Methods:
A novel series of 3-[4-(2-amino-5, 6-dihydro-4(substituted phenyl)-4H-1, 3-oxazin
/thiazin-6yl) phenyl]-2, 3-dihyro-2-thioxoquinazolin-4(1H)-one derivatives (4a-4p) were synthesized.
The structures of the synthesized compounds were assigned on the basis of spectral data (UV,
IR, 1HNMR, 13CNMR and MS) and performed anticonvulsant activity against maximal electroshock
test and Subcutaneous Pentylenetetrazole model. Neurotoxicity was assessed using a rotarod apparatus
test. The molecular docking study was performed to assess their binding affinities towards
Gamma-Aminobutyric Acid type A receptor. A quantitative estimate of drug-likeness was also performed,
which calculates the molecular properties and screen the molecules based on drug-likeness
rules.
Results:
Compounds 4b, 4e, 4j and 4m have shown the highest anticonvulsant activity against tonic
seizure with decreased mean duration of tonic hind leg extension of 8.31, 7.35, 8.61 and 8.99 s, respectively
in maximal electroshock model and increased onset time clonic convulsion duration of
94.45, 96.65, 93.51 and 91.86 s in Subcutaneous Pentylenetetrazole model. Molecular docking study
revealed a better binding affinity with Gamma-Aminobutyric Acid type A receptor.
Conclusion:
The compound 4b and 4e emerged out as the pilot molecule with a better anticonvulsant
activity without any neurotoxicity. The obtained results showed that compounds 4b and 4e
could be useful as a template for future design, optimization, and investigation to produce more active
analogs.
Collapse
Affiliation(s)
- Nimisha jain
- Computational & Synthetic Chemistry Division, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, VNS Group of Institutions, Neelbud, Bhopal 462023 (M.P.), India
| | - Pradeep Kumar Singour
- Computational & Synthetic Chemistry Division, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, VNS Group of Institutions, Neelbud, Bhopal 462023 (M.P.), India
| |
Collapse
|
27
|
Jain N, Singour PK. Novel 3-{4-[2-Amino-4-(Substitutedphenyl)-2H-[1, 3] Oxazin/Thiazin-6-Yl} -2-Phenyl-3H-Quinazolin-4-One Derivatives as Enhancer of GABA Mediated Inhibition: Synthesis, Molecular Modeling and Pharmacological Studies. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190222155404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background:
According to WHO, the 50 million people worldwide are suffering from
epilepsy, making it one of the most common neurological diseases globally. Epilepsy is often
characterized by neurobiological, cognitive, psychological and behavioral changes and that may
enhance the susceptibility to seizures and affect the quality of life.
Objective:
The aim of the present work was to develop 2, 3 disubstituted 4-(3H)-quinazolinone
derivatives in order to find an effective and highly lipophilic compound with lesser side effects and
to evaluate them for anticonvulsant and neurotoxic activity.
Methods:
A novel series of 3-4-[2-amino-4-(substitutedphenyl)-2H-[1.3] oxazin/thiazin-6-yl 2-
phenyl-3H-quinazolin-4-one derivatives were synthesized and evaluated for their anticonvulsant
activity. The structures of the compound have been confirmed by spectral analysis. The molecular
docking study was performed for finding the binding affinity with GABAA receptor in order to
rationalize their anticonvulsant activities in a qualitative way. Quantitative estimate of drug-likeness
was also performed which calculate the molecular properties and screen the molecules based on
drug-likeness rules. Anticonvulsant activities of synthesized compounds were done by using
(Maximal electroshock) MES induced seizures and subcutaneous pentylenetetrazole (scPTZ) induced
seizure models in Wistar rats of either sex. None of the compounds demonstrated any sign of
neurotoxicity.
Results:
Compounds 3-4-[2-amino-4-(fluorophenyl)-2H- [1, 3] oxazin-6-yl 2-phenyl-3H
quinazolin-4-one (5i) and 3-4-[2-amino-4-(fluorophenyl)-2H- [1, 3] thiazin -6-yl 2-phenyl-3H
quinazolin-4-one (5n) have shown significant activity against tonic seizure by the MES model and
clonic seizure by scPTZ induced seizure model.
Conclusion:
These ten novels synthesized compounds had significant anticonvulsant activity. As a
result, the compound (5i) and (5n) emerged out as the pilot molecule with a better anticonvulsant
activity without any neurotoxicity, while the other compounds have moderate activity. QED analysis
of compounds (5i) and (5n) also indicated that these compounds will have good oral absorption. The
proposed work is to make efforts towards the development and identification of novel molecules as
anticonvulsant agents by the synthesis of some novel quinazolinone derivatives with improved
biological activity.
Collapse
Affiliation(s)
- Nimisha Jain
- Department of Pharmaceutical Chemistry, Computational and Synthetic Chemistry Division, Faculty of Pharmacy,VNS Group of Institutions, Neelbud, Bhopal 462023 (M.P.), India
| | - Pradeep Kumar Singour
- Department of Pharmaceutical Chemistry, Computational and Synthetic Chemistry Division, Faculty of Pharmacy,VNS Group of Institutions, Neelbud, Bhopal 462023 (M.P.), India
| |
Collapse
|
28
|
Dowarah J, Singh VP. Anti-diabetic drugs recent approaches and advancements. Bioorg Med Chem 2020; 28:115263. [PMID: 32008883 DOI: 10.1016/j.bmc.2019.115263] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/20/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Diabetes is one of the major diseases worldwide and is the third leading cause of death in the United States. Anti-diabetic drugs are used in the treatment of diabetes mellitus to control glucose levels in the blood. Most of the drugs are administered orally, except for a few of them, such as insulin, exenatide, and pramlintide. In this review, we are going to discuss seven major types of anti-diabetic drugs: Peroxisome proliferator-activated receptor (PPAR) agonist, protein tyrosine phosphatase 1B (PTP1B) inhibitors, aldose reductase inhibitors, α-glucosidase inhibitors, dipeptidyl peptidase IV (DPP-4) inhibitors, G protein-coupled receptor (GPCR) agonists and sodium-glucose co-transporter (SGLT) inhibitors. Here, we are also discussing some of the recently reported anti-diabetic agents with its multi-target pharmacological actions. This review summarises recent approaches and advancement in anti-diabetes treatment concerning characteristics, structure-activity relationships, functional mechanisms, expression regulation, and applications in medicine.
Collapse
Affiliation(s)
- Jayanta Dowarah
- Department of Chemistry, Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | - Ved Prakash Singh
- Department of Chemistry, Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India.
| |
Collapse
|
29
|
Rafique R, Khan KM, Arshia, Chigurupati S, Wadood A, Rehman AU, Salar U, Venugopal V, Shamim S, Taha M, Perveen S. Synthesis, in vitro α-amylase inhibitory, and radicals (DPPH & ABTS) scavenging potentials of new N-sulfonohydrazide substituted indazoles. Bioorg Chem 2020; 94:103410. [PMID: 31732193 DOI: 10.1016/j.bioorg.2019.103410] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 11/22/2022]
Abstract
Over-expression of α-amylase enzyme causes hyperglycemia which lead to many physiological complications including oxidative stress, one of the most commonly associated problem with diabetes mellitus. Marketed α-amylase inhibitors such as acarbose, voglibose, and miglitol used to treat type-II diabetes mellitus, but also linked to several harmful effects. Therefore, it is essential to explore new and nontoxic antidiabetic agents with additional antioxidant properties. In this connection, a series of new N-sulfonohydrazide substituted indazoles 1-19 were synthesized by multistep reaction scheme and assessed for in vitro α-amylase inhibitory and radical (DPPH and ABTS) scavenging properties. All compounds were fully characterized by different spectroscopic techniques including 1H, 13C NMR, EI-MS, HREI-MS, ESI-MS, and HRESI-MS. Compounds showed promising α-amylase inhibitory activities (IC50 = 1.23 ± 0.06-4.5 ± 0.03 µM) as compared to the standard acarbose (IC50 1.20 ± 0.09 µM). In addition to that all derivatives were found good to moderate scavengers of DPPH (IC50 2.01 ± 0.13-5.3 ± 0.11) and ABTS (IC50 = 2.34 ± 0.07-5.5 ± 0.07 µM) radicals, in comparison with standard ascorbic acid having scavenging activities with IC50 = 1.99 ± 0.09 µM, and IC50 2.03 ± 0.11 µM for DPPH and ABTS radicals. In silico molecular docking study was conducted to rationalize the binding interaction of α-amylase enzyme with ligands. Compounds were observed as mixed type inhibitors in enzyme kinetic characterization.
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
| | - 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.
| | - Arshia
- 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, Faculty 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
| | - Uzma Salar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | | | - Shahbaz Shamim
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological, Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Karachi, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi 75280, Pakistan
| |
Collapse
|
30
|
Rafique R, Khan KM, Arshia, Kanwal, Chigurupati S, Wadood A, Rehman AU, Karunanidhi A, Hameed S, Taha M, Al-Rashida M. Synthesis of new indazole based dual inhibitors of α-glucosidase and α-amylase enzymes, their in vitro, in silico and kinetics studies. Bioorg Chem 2020; 94:103195. [PMID: 31451297 DOI: 10.1016/j.bioorg.2019.103195] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/01/2019] [Accepted: 08/11/2019] [Indexed: 12/20/2022]
Abstract
The current study describes the discovery of novel inhibitors of α-glucosidase and α-amylase enzymes. For that purpose, new hybrid analogs of N-hydrazinecarbothioamide substituted indazoles 4-18 were synthesized and fully characterized by EI-MS, FAB-MS, HRFAB-MS, 1H-, and 13C NMR spectroscopic techniques. Stereochemistry of the imine double bond was established by NOESY measurements. All derivatives 4-18 with their intermediates 1-3, were evaluated for in vitro α-glucosidase and α-amylase enzyme inhibition. It is worth mentioning that all synthetic compounds showed good inhibition potential in the range of 1.54 ± 0.02-4.89 ± 0.02 µM for α-glucosidase and for α-amylase 1.42 ± 0.04-4.5 ± 0.18 µM in comparison with the standard acarbose (IC50 value of 1.36 ± 0.01 µM). In silico studies were carried out to rationalize the mode of binding interaction of ligands with the active site of enzymes. Moreover, enzyme inhibitory kinetic characterization was also performed to understand the mechanism of enzyme inhibition.
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
| | - 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.
| | - 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
| | - Sridevi Chigurupati
- Department of Pharmacology, Faculty 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
| | - Arunkumar Karunanidhi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Shehryar Hameed
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Lahore, Pakistan
| |
Collapse
|
31
|
Auti PS, George G, Paul AT. Recent advances in the pharmacological diversification of quinazoline/quinazolinone hybrids. RSC Adv 2020; 10:41353-41392. [PMID: 35516563 PMCID: PMC9057921 DOI: 10.1039/d0ra06642g] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Due to the pharmacological activities of quinazoline and quinazolinone scaffolds, it has aroused great interest in medicinal chemists for the development of new drugs or drug candidates. The pharmacological activities of quinazoline and its related scaffolds include anti-cancer, anti-microbial, anti-convulsant, and antihyperlipidaemia. Recently, molecular hybridization technology is used for the development of hybrid analogues with improved potency by combining two or more pharmacophores of bioactive scaffolds. The molecular hybridization of various biologically active pharmacophores with quinazoline derivatives resulted in lead compounds with multi-faceted biological activity wherein specific as well as multiple targets were involved. The present review summarizes the advances in lead compounds of quinazoline hybrids and their related heterocycles in medicinal chemistry. Moreover, the review also helps to intensify the drug development process by providing an understanding of the potential role of these hybridized pharmacophoric features in exhibiting various pharmacological activities. Recent advances in quinazoline/quinazolinone hybrid heterocycles in medicinal chemistry and their pharmacological diversification.![]()
Collapse
Affiliation(s)
- Prashant S. Auti
- Laboratory of Natural Product Chemistry
- Department of Pharmacy
- Birla Institute of Technology and Science, Pilani (BITS Pilani)
- Pilani Campus
- India
| | - Ginson George
- Laboratory of Natural Product Chemistry
- Department of Pharmacy
- Birla Institute of Technology and Science, Pilani (BITS Pilani)
- Pilani Campus
- India
| | - Atish T. Paul
- Laboratory of Natural Product Chemistry
- Department of Pharmacy
- Birla Institute of Technology and Science, Pilani (BITS Pilani)
- Pilani Campus
- India
| |
Collapse
|
32
|
Lopéz D, Cherigo L, Mejia LC, Loza-Mejía MA, Martínez-Luis S. α-Glucosidase inhibitors from a mangrove associated fungus, Zasmidium sp. strain EM5-10. BMC Chem 2019; 13:22. [PMID: 31384771 PMCID: PMC6661824 DOI: 10.1186/s13065-019-0540-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/29/2019] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Mangroves plants and their endophytes represent a natural source of novel and bioactive compounds. In our ongoing research on mangrove endophytes from the Panamanian Pacific Coast, we have identified several bioactive endophytic fungi. From these organisms, an isolate belonging to the genus Zasmidium (Mycosphaerellaceae) showed 91.3% of inhibition against α-glucosidase enzyme in vitro. RESULTS Zasmidium sp. strain EM5-10 was isolated from mature leaves of Laguncularia racemosa, and its crude extract showed good inhibition against α-glucosidase enzyme (91.3% of inhibition). Bioassay-guided fractionation of the crude extract led to obtaining two active fractions: L (tripalmitin) and M (Fungal Tryglicerides Mixture). Tripalmitin (3.75 µM) showed better inhibitory activity than acarbose (positive control, IC50 217.71 µM). Kinetic analysis established that tripalmitin acted as a mixed inhibitor. Molecular docking and molecular dynamics simulations predicted that tripalmitin binds at the same site as acarbose and also to an allosteric site in the human intestinal α-glucosidase (PDB: 3TOP). CONCLUSIONS Zasmidium sp. strain EM5-10 represents a new source of bioactive substances that could possess beneficial properties for human health.
Collapse
Affiliation(s)
- Dioxelis Lopéz
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Edificio 208, Ciudad del Saber, Apartado, 0843-01103 Panama City, Panama
- Department of Biotechnology, Acharya Nagarjuna University, Nagarjuna Nagar, Guntur, 522510 India
| | - Lilia Cherigo
- Departamento de Química Orgánica, Escuela De Química, Facultad de Ciencias Exactas y Tecnología, Universidad de Panamá, P.O. Box 3366, Panama City, Panama
| | - Luis C. Mejia
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Edificio 208, Ciudad del Saber, Apartado, 0843-01103 Panama City, Panama
| | - Marco A. Loza-Mejía
- Facultad de Ciencias Químicas, Universidad La Salle, Benjamín Franklin 45, Cuauhtémoc, 06140 Mexico City, Mexico
| | - Sergio Martínez-Luis
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Edificio 208, Ciudad del Saber, Apartado, 0843-01103 Panama City, Panama
| |
Collapse
|
33
|
Zhang D, Shi Y, Li J, Ruan D, Jia Q, Zhu W, Chen K, Li Y, Wang R. Alkaloids with Nitric Oxide Inhibitory Activities from the Roots of Isatis tinctoria. Molecules 2019; 24:molecules24224033. [PMID: 31703370 PMCID: PMC6891263 DOI: 10.3390/molecules24224033] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022] Open
Abstract
As our ongoing research project on Ban Lan Gen (Isatis tinctoria roots), a total of 23 alkaloids were obtained. Compounds 1 and 2 contain an unusual C–C bond between the 2(1H)-quinolinone moiety and the phenol moiety and between the 2(1H)-quinolinone moiety and the 1H-indole moiety, respectively. Compound 3 possesses an unusual carbon skeleton and its putative biosynthetic pathway was discussed, and Compound 23 was deduced as a new indole alkaloid glycoside. Compounds 4–7 were identified as four new natural products by extensive spectroscopic experiments. Additionally, the anti-inflammatory activity was assessed based on nitric oxide (NO) production using Lipopolysaccharide-stimulated RAW264.7 macrophages. Compounds 9, 15, and 17 showed inhibitory effects with IC50 values of 1.2, 5.0, and 74.4 μM.
Collapse
Affiliation(s)
- Dongdong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
| | - Yanhong Shi
- Institute of TCM International Standardization of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Jingyi Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
| | - Deqing Ruan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
| | - Qi Jia
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
| | - Weiliang Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China;
| | - Kaixian Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China;
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
- Correspondence: (Y.L.); (R.W.); Tel.: +86-21-5132-2191 (Y.L.); +86-21-5132-2181 (R.W.); Fax: +86-21-5132-2193 (Y.L. & R.W.)
| | - Rui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (D.Z.); (J.L.); (D.R.); (Q.J.); (K.C.)
- Correspondence: (Y.L.); (R.W.); Tel.: +86-21-5132-2191 (Y.L.); +86-21-5132-2181 (R.W.); Fax: +86-21-5132-2193 (Y.L. & R.W.)
| |
Collapse
|
34
|
Hussen AS, Bagchi S, Sharma A. Ammonium Chloride Assisted Microwave Mediated Domino Multicomponent Reaction: An Efficient and Sustainable Synthesis of Quinazolin‐4(3
H
)‐imines under Solvent Free Condition. ChemistrySelect 2019. [DOI: 10.1002/slct.201900923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
| | - Sourav Bagchi
- Department of ChemistryIndian Institute of Technology Roorkee Roorkee- 247667
| | - Anuj Sharma
- Department of ChemistryIndian Institute of Technology Roorkee Roorkee- 247667
| |
Collapse
|
35
|
Ullah S, Mirza S, Salar U, Hussain S, Javaid K, Khan KM, Khalil R, Atia-Tul-Wahab, Ul-Haq Z, Perveen S, Choudhary MI. 2-Mercapto Benzothiazole Derivatives: As Potential Leads for the Diabetic Management. Med Chem 2019; 16:826-840. [PMID: 31195949 DOI: 10.2174/1573406415666190612153150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 05/07/2019] [Accepted: 05/17/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Results of our previous studies on antiglycation activity, and the noncytotoxicity of 2-mercapto benzothiazoles, encouraged us to further widen our investigation towards the identification of leads against diabetes mellitus. METHODS 33 derivatives of 2-mercapto benzothiazoles 1-33 were evaluated for in vitro α- glucosidase inhibitory activity. Mode of inhibition was deduced by kinetic studies. To predict the interactions of 2-mercapto benzothiazole derivatives 1-33 with the binding pocket of α-glucosidase enzyme, molecular docking studies were performed on the selected inhibitors. RESULTS Compounds 2-4, 6-7, 9-26, 28 and 30 showed many folds potent α-glucosidase inhibitory activity in the range of IC50 = 31.21-208.63 μM, as compared to the standard drug acarbose (IC50 = 875.75 ± 2.08 μM). It was important to note that except derivative 28, all other derivatives were also found previously to have antiglycating potential in the range of IC50 = 187.12-707.21 μM. CONCLUSION A number of compounds were identified as dual nature as antiglycating agent and α- glucosidase inhibitors. These compounds may serve as potential lead candidates for the management of diabetes mellitus.
Collapse
Affiliation(s)
- Saeed Ullah
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Salma Mirza
- 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
| | - Shafqat Hussain
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Kulsoom Javaid
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Khalid M 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
| | - Ruqaiya Khalil
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Atia-Tul-Wahab
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Shahnaz Perveen
- PCSIR Laboratories Complex Karachi, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi-75280, Pakistan
| | - Muhammad I Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan,Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah-21412, Saudi Arabia
| |
Collapse
|
36
|
Design and synthesis of novel quinazolinone-1,2,3-triazole hybrids as new anti-diabetic agents: In vitro α-glucosidase inhibition, kinetic, and docking study. Bioorg Chem 2019; 83:161-169. [DOI: 10.1016/j.bioorg.2018.10.023] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 01/13/2023]
|
37
|
Nur-e-Alam M, Yousaf M, Parveen I, Hafizur RM, Ghani U, Ahmed S, Hameed A, Threadgill MD, Al-Rehaily AJ. New flavonoids from the Saudi Arabian plant Retama raetam which stimulates secretion of insulin and inhibits α-glucosidase. Org Biomol Chem 2019; 17:1266-1276. [DOI: 10.1039/c8ob02755b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Retama raetam is a bush which is used in traditional medicine.
Collapse
Affiliation(s)
- Mohammad Nur-e-Alam
- Department of Pharmacognosy
- College of Pharmacy
- King Saud University
- Riyadh 11451
- Kingdom of Saudi Arabia
| | - Muhammad Yousaf
- Department of Pharmacognosy
- College of Pharmacy
- King Saud University
- Riyadh 11451
- Kingdom of Saudi Arabia
| | - Ifat Parveen
- Institute of Biological
- Environmental & Rural Sciences (IBERS)
- Aberystwyth University
- Aberystwyth SY23 3DA
- UK
| | - Rahman M. Hafizur
- Dr. Panjwani Center for Molecular Medicine and Drug Research
- International Center for Chemical and Biological Sciences
- University of Karachi
- Karachi-75270
- Pakistan
| | - Usman Ghani
- Clinical Biochemistry Unit
- Department of Pathology
- College of Medicine
- King Saud University
- Riyadh 11461
| | - Sarfaraz Ahmed
- Department of Pharmacognosy
- College of Pharmacy
- King Saud University
- Riyadh 11451
- Kingdom of Saudi Arabia
| | - Abdul Hameed
- Dr. Panjwani Center for Molecular Medicine and Drug Research
- International Center for Chemical and Biological Sciences
- University of Karachi
- Karachi-75270
- Pakistan
| | - Michael D. Threadgill
- Institute of Biological
- Environmental & Rural Sciences (IBERS)
- Aberystwyth University
- Aberystwyth SY23 3DA
- UK
| | - Adnan J. Al-Rehaily
- Department of Pharmacognosy
- College of Pharmacy
- King Saud University
- Riyadh 11451
- Kingdom of Saudi Arabia
| |
Collapse
|
38
|
Kazmi M, Zaib S, Ibrar A, Amjad ST, Shafique Z, Mehsud S, Saeed A, Iqbal J, Khan I. A new entry into the portfolio of α-glucosidase inhibitors as potent therapeutics for type 2 diabetes: Design, bioevaluation and one-pot multi-component synthesis of diamine-bridged coumarinyl oxadiazole conjugates. Bioorg Chem 2018; 77:190-202. [DOI: 10.1016/j.bioorg.2017.12.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 10/30/2017] [Accepted: 12/20/2017] [Indexed: 01/15/2023]
|
39
|
Nagamoto M, Yorimitsu H, Nishimura T. Iridium-Catalyzed Hydroarylation of Conjugated Dienes via π-Allyliridium Intermediates. Org Lett 2018; 20:828-831. [DOI: 10.1021/acs.orglett.7b03969] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Midori Nagamoto
- Department
of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Hideki Yorimitsu
- Department
of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Takahiro Nishimura
- Department
of Chemistry, Graduate School of Science, Osaka City University, Sumiyoshi, Osaka 558-8585, Japan
| |
Collapse
|
40
|
Salar U, Khan KM, Chigurupati S, Taha M, Wadood A, Vijayabalan S, Ghufran M, Perveen S. New Hybrid Hydrazinyl Thiazole Substituted Chromones: As Potential α-Amylase Inhibitors and Radical (DPPH & ABTS) Scavengers. Sci Rep 2017; 7:16980. [PMID: 29209017 PMCID: PMC5717224 DOI: 10.1038/s41598-017-17261-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022] Open
Abstract
Current research is based on the identification of novel inhibitors of α-amylase enzyme. For that purpose, new hybrid molecules of hydrazinyl thiazole substituted chromones 5-27 were synthesized by multi-step reaction and fully characterized by various spectroscopic techniques such as EI-MS, HREI-MS, 1H-NMR and 13C-NMR. Stereochemistry of the iminic bond was confirmed by NOESY analysis of a representative molecule. All compounds 5-27 along with their intervening intermediates 1-4, were screened for in vitro α-amylase inhibitory, DPPH and ABTS radical scavenging activities. All compounds showed good inhibition potential in the range of IC50 = 2.186-3.405 µM as compared to standard acarbose having IC50 value of 1.9 ± 0.07 µM. It is worth mentioning that compounds were also demonstrated good DPPH (IC50 = 0.09-2.233 µM) and ABTS (IC50 = 0.584-3.738 µM) radical scavenging activities as compared to standard ascorbic acid having IC50 = 0.33 ± 0.18 µM for DPPH and IC50 = 0.53 ± 0.3 µM for ABTS radical scavenging activities. In addition to that cytotoxicity of the compounds were checked on NIH-3T3 mouse fibroblast cell line and found to be non-toxic. In silico studies were performed to rationalize the binding mode of compounds (ligands) with the active site of α-amylase enzyme.
Collapse
Affiliation(s)
- Uzma Salar
- 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 Pharmaceutical chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100, Bedong, Kedah, Malaysia
| | - 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
| | - Abdul Wadood
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, UCSS, Abdul Wali Khan University, Mardan, Pakistan
| | - Shantini Vijayabalan
- Department of Pharmaceutical chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100, Bedong, Kedah, Malaysia
| | - Mehreen Ghufran
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, UCSS, Abdul Wali Khan University, Mardan, Pakistan
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Karachi, Shahrah-e-Dr. SalimuzzamanSiddiqui, Karachi, 75280, Pakistan
| |
Collapse
|
41
|
Luthra T, Agarwal R, Estari M, Adepally U, Sen S. A novel library of -arylketones as potential inhibitors of α-glucosidase: Their design, synthesis, in vitro and in vivo studies. Sci Rep 2017; 7:13246. [PMID: 29038580 PMCID: PMC5643545 DOI: 10.1038/s41598-017-13798-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/26/2017] [Indexed: 11/09/2022] Open
Abstract
α-glucosidase is an essential enzyme located at the brush border of intestines. It is an important therapeutic target for type II diabetes. Herein we have designed a library of novel α-arylketones as inhibitors of α-glucosidase (yeast origin) via scaffold hopping and bioisosteric modification of known inhibitors of α-glucosidase. The design was validated through molecular docking that revealed strong binding interactions of the newly designed compounds against α-glucosidase. A library comprising of 15 compounds was synthesized in a combinatorial fashion, where the advanced amide intermediates were accessed through “shot gun” synthesis. The final compounds were characterized by 1H, 13C-NMR and with high resolution mass spectroscopy. In vitro screening of the compounds against yeast α-glucosidase revealed substantial inhibition with IC50s in the range of 4–10 μM (the standard drug acarbose inhibits α-glucosidase with an IC50 of 9.95 μM). Reaction kinetics suggested mixed type inhibition. Finally, in vivo studies of the most active compound 3c against Streptozotocin induced male albino Wistar rats revealed that its administration in the rats for about 4 weeks lead to a highly significant (P < 0.001) decrease in the fasting blood glucose (FBG) compared to the untreated diabetic rats. Moreover, lower dose of 3c had better control over FBG in contrast to high-dose.
Collapse
Affiliation(s)
- Tania Luthra
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Dadri, Chithera, GautamBudh Nagar, Uttar Pradesh, 201314, India
| | - Rahul Agarwal
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Chithera, GautamBudh Nagar, Uttar Pradesh, 201314, India
| | - Mamidala Estari
- Department of Zoology, Kakatiya University, Warangal-506009, Telengana, India
| | - Uma Adepally
- Institute of Science and Technology Jawaharlal Nehru Technological University, Kukatpally, Hyderabad, Telangana, India.
| | - Subhabrata Sen
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Dadri, Chithera, GautamBudh Nagar, Uttar Pradesh, 201314, India.
| |
Collapse
|
42
|
Kazmi M, Zaib S, Amjad ST, Khan I, Ibrar A, Saeed A, Iqbal J. Exploration of aroyl/heteroaroyl iminothiazolines featuring 2,4,5-trichlorophenyl moiety as a new class of potent, selective, and in vitro efficacious glucosidase inhibitors. Bioorg Chem 2017; 74:134-144. [DOI: 10.1016/j.bioorg.2017.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/16/2017] [Accepted: 07/25/2017] [Indexed: 12/29/2022]
|
43
|
Devi J, Kalita SJ, Deka DC. Expeditious synthesis of 2,3-dihydroquinazolin-4(1H)-ones in aqueous medium using thiamine hydrochloride (VB1) as a mild, efficient, and reusable organocatalyst. SYNTHETIC COMMUN 2017. [DOI: 10.1080/00397911.2017.1337149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jutika Devi
- Department of Chemistry, University of Gauhati, Guwahati, Assam, India
| | | | | |
Collapse
|
44
|
Liu Z, Ma S. Recent Advances in Synthetic α-Glucosidase Inhibitors. ChemMedChem 2017; 12:819-829. [PMID: 28498640 DOI: 10.1002/cmdc.201700216] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/08/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Zhiyang Liu
- Department of Medicinal Chemistry; Key Laboratory of Chemical Biology (Ministry of Education); School of Pharmaceutical Sciences; Shandong University; 44 West Culture Road Jinan 250012 P.R. China
| | - Shutao Ma
- Department of Medicinal Chemistry; Key Laboratory of Chemical Biology (Ministry of Education); School of Pharmaceutical Sciences; Shandong University; 44 West Culture Road Jinan 250012 P.R. China
| |
Collapse
|
45
|
Wei M, Chai WM, Wang R, Yang Q, Deng Z, Peng Y. Quinazolinone derivatives: Synthesis and comparison of inhibitory mechanisms on α-glucosidase. Bioorg Med Chem 2017; 25:1303-1308. [DOI: 10.1016/j.bmc.2016.09.042] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 12/21/2022]
|
46
|
|
47
|
Wang R, Chai WM, Yang Q, Wei MK, Peng Y. 2-(4-Fluorophenyl)-quinazolin-4(3H)-one as a novel tyrosinase inhibitor: Synthesis, inhibitory activity, and mechanism. Bioorg Med Chem 2016; 24:4620-4625. [PMID: 27527415 DOI: 10.1016/j.bmc.2016.07.068] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/26/2016] [Accepted: 07/30/2016] [Indexed: 01/12/2023]
Abstract
2-(4-Fluorophenyl)-quinazolin-4(3H)-one (FQ) was synthesized, and its structure was identified with (1)H nuclear magnetic resonance ((1)H NMR), (13)C nuclear magnetic resonance ((13)C NMR), fourier transform infrared spectroscopy (FTIR), and high resolution mass spectrometry (HRMS). From the enzyme analysis, the results showed that it could inhibit the diphenolase activity of tyrosinase (IC50=120±2μM). Furthermore, the results of kinetic studies showed that the compound was a reversible mixed-type inhibitor, and that the inhibition constants were determined to be 703.2 (KI) and 222.1μM (KIS). The results of fluorescence quenching experiment showed that the compound could interact with tyrosinase and the substrates (tyrosine and l-DOPA). Molecular docking analysis revealed that the mass transfer rate was affected by FQ blocking the enzyme catalytic center. In brief, current study identified a novel tyrosinase inhibitor which deserved further study for hyperpigmentation drugs.
Collapse
Affiliation(s)
- Rui Wang
- Key Laboratory of Small Fuctional Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Wei-Ming Chai
- Key Laboratory of Small Fuctional Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi 330022, China; Key Laboratory of Green Chemistry, Nanchang, Jiangxi 330022, China.
| | - Qin Yang
- Key Laboratory of Small Fuctional Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi 330022, China; Key Laboratory of Green Chemistry, Nanchang, Jiangxi 330022, China
| | - Man-Kun Wei
- Key Laboratory of Small Fuctional Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Yiyuan Peng
- Key Laboratory of Small Fuctional Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi 330022, China; Key Laboratory of Green Chemistry, Nanchang, Jiangxi 330022, China.
| |
Collapse
|
48
|
Khan I, Zaib S, Batool S, Abbas N, Ashraf Z, Iqbal J, Saeed A. Quinazolines and quinazolinones as ubiquitous structural fragments in medicinal chemistry: An update on the development of synthetic methods and pharmacological diversification. Bioorg Med Chem 2016; 24:2361-2381. [PMID: 27112448 DOI: 10.1016/j.bmc.2016.03.031] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/16/2016] [Accepted: 03/18/2016] [Indexed: 12/17/2022]
Abstract
Nitrogen-rich heterocycles, particularly quinazolines and quinazolinones, represent a unique class of diversified frameworks displaying a broad spectrum of biological functions. Over the past several years, intensive medicinal chemistry efforts have generated numerous structurally functionalized quinazoline and quinazolinone derivatives. Interest in expanding the biological effects, demonstrated by these motifs, is growing exponentially, as indicated by the large number of publications reporting the easy accessibility of these skeletons in addition to the diverse nature of synthetic as well as biological applications. Therefore, the main focus of the present review is to provide an ample but condensed overview on various synthetic approaches providing access to quinazoline and quinazolinone compounds with multifaceted biological activities. Furthermore, mechanistic insights, synthetic utilization, structure-activity relationships and molecular modeling inputs for the potent derivatives have also been discussed.
Collapse
Affiliation(s)
- Imtiaz Khan
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| | - Sumera Zaib
- Department of Biochemistry, Hazara University, Garden Campus, Mansehra, Pakistan; Centre for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Sadaf Batool
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad 44000, Pakistan
| | - Naeem Abbas
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Zaman Ashraf
- Department of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
49
|
Synthesis and biological evaluation of heterocyclic privileged medicinal structures containing (benz)imidazole unit. MONATSHEFTE FUR CHEMIE 2016. [DOI: 10.1007/s00706-016-1733-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|