1
|
Wang YH, Liu TW, Hsiao SW, Chu MH, Lee TH, Hsu SJ, Chen SY, Lee CK. Comparative metabolomics of acetylcholinesterase and α-glucosidase inhibitors in pericarp of Garcinia mangostana L. BOTANICAL STUDIES 2025; 66:13. [PMID: 40397045 PMCID: PMC12095715 DOI: 10.1186/s40529-025-00460-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/10/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Mangosteen (Garcinia mangostana L.) pericarp extract has demonstrated potential against Alzheimer's disease (AD) and diabetes mellitus (DM). This study introduces a rapid dereplication and comparative approach to identify and characterize acetylcholinesterase (AChE) and α-glucosidase inhibitors in mangosteen pericarp. Using protein-subtraction, MS profiling, and computational modeling is effective for screening, identifying, and analyzing enzyme-inhibiting compounds from plant sources, and quantitative analysis of the main components has been performed. RESULTS The Mangosteen pericarp extract observed significant inhibitory activity against α-glucosidase and AChE, with IC50 values of 31.02 and 70.56 µg/mL, respectively. By comparing profiles of protein-subtracted extracts with non-treated extracts, eight potential inhibitors for each enzyme were identified: 8-desoxygartanin, gartanin, 3-isomangostin, β-mangostin, 9-hydroxycalabaxanthone, γ-mangostin, α-mangostin, and garcinone E. The α-mangostin was the most abundant, comprising 39.589% of the extract. Molecular docking revealed these inhibitors target the peripheral anionic site of AChE and the active site of α-glucosidase, forming key hydrogen bonds and pi-pi stacking interactions. CONCLUSION This study emphasizes mangosteen pericarp as a promising natural source of these inhibitors, with potential for use in developing nutraceuticals and pharmaceuticals. The study validated a systems biology approach by applying dereplication and comparative UPLC-ESI-MS/MS metabolomics profiling to identify target-binding molecules in both protein-treated and untreated plant extracts. Further confirmation was obtained through molecular docking predictions, mechanism analysis, and compound quantification assays.
Collapse
Affiliation(s)
- Yun-Han Wang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, 250 Wu Xin Street, Taipei, 110, Taiwan
| | - Ta-Wei Liu
- School of Pharmacy, Taipei Medical University, 250 Wu Xin Street, Taipei, 110, Taiwan
| | - Sui-Wen Hsiao
- Ph.D. Program in Drug Discovery and Development Industry, Taipei Medical University, 250 Wu Xin Street, Taipei, 11031, Taiwan
| | - Man-Hsiu Chu
- School of Pharmacy, Taipei Medical University, 250 Wu Xin Street, Taipei, 110, Taiwan
| | - Tzong-Huei Lee
- Institute of Fisheries Science, National Taiwan University, Taipei City, Taiwan
| | - Su-Jung Hsu
- School of Pharmacy, Taipei Medical University, 250 Wu Xin Street, Taipei, 110, Taiwan.
- Institute of Fisheries Science, National Taiwan University, Taipei City, Taiwan.
| | - Shih Yin Chen
- Xantho Biotechnology Co., Ltd. Rm ER15, 17F.-1, No. 3, Yuanqu St., Nangang Dist., Taipei City, 11503, Taiwan (R.O.C.)
| | - Ching-Kuo Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, 250 Wu Xin Street, Taipei, 110, Taiwan.
- School of Pharmacy, Taipei Medical University, 250 Wu Xin Street, Taipei, 110, Taiwan.
- Ph.D. Program in Drug Discovery and Development Industry, Taipei Medical University, 250 Wu Xin Street, Taipei, 11031, Taiwan.
- Graduate Institute of Pharmacognosy, Taipei Medical University, 250 Wu Xin Street, Taipei, 11031, Taiwan.
- School of Food Safety, Taipei Medical University, 250 Wu Xin Street, Taipei, 11031, Taiwan.
- Department of Chemistry, Chung Yuan Christian University, 200 Zhongbei Road, Zhongli District, Taoyuan, 32023, Taiwan.
| |
Collapse
|
2
|
Naeem N, Zaib S, Mughal EU, Othman GA, Sadiq A, Rana N. Discovery of bis-chalcones and bis-pyrimidines as potential urease inhibitors: from synthesis to computational insights. Future Med Chem 2025; 17:929-941. [PMID: 40183169 DOI: 10.1080/17568919.2025.2485870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
AIMS This study focuses on the design and evaluation of bis-chalcones and bis-pyrimidines as potential urease inhibitors. MATERIALS AND METHODS A series of bis-chalcone and bis-pyrimidine derivatives were synthesized and assessed for their in vitro urease inhibitory activity. Kinetic studies were conducted using Lineweaver-Burk plots to determine the inhibition mechanism of the most potent compound. Molecular docking was employed to investigate the binding interactions with the urease active site, followed by MD simulations to validate complex stability. Computational ADMET analysis was performed to assess the drug-like properties of the most active inhibitor. RESULTS Several synthesized compounds exhibited potent urease inhibitory activity, significantly surpassing the standard inhibitor thiourea. The most active compound, 8P, displayed noncompetitive inhibition, as confirmed by kinetic studies. SAR analysis revealed that electron-withdrawing substituents enhanced inhibitory potency. Molecular docking studies demonstrated favorable interactions between inhibitors and key urease residues, while MD simulations confirmed complex stability. ADMET analysis supported the drug-like potential of 8P. CONCLUSIONS This study provides valuable insights into the development of target compounds as promising urease inhibitors. These findings suggest their potential therapeutic applications for urease-related disorders.
Collapse
Affiliation(s)
- Nafeesa Naeem
- Department of Chemistry, University of Gujrat, Gujrat, Pakistan
| | - Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | | | - Gehan Ahmed Othman
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Amina Sadiq
- Department of Chemistry, Government College Women University, Sialkot, Pakistan
| | - Nehal Rana
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
3
|
Chen YT, Wan BW, Wang KM, Zhu KK, Meng N, Jiang CS, Zhang J. Design and synthesis of N-(3-cyanothiophen-2-yl)-2-phenoxyacetamide-based α-glucosidase inhibitors. Bioorg Med Chem Lett 2025; 117:130068. [PMID: 39662707 DOI: 10.1016/j.bmcl.2024.130068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/17/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
This study investigates the design and synthesis of a series of novel selective α-glucosidase inhibitors based on N-(3-cyanothiophen-2-yl)-2-phenoxyacetamide framework, employing a bioisosterism strategy. Among the nineteen newly synthesized analogs, compound 4d9 demonstrated the highest α-glucosidase inhibitory potency (IC50 = 2.11 μM) when compared to the established inhibitors Acarbose (IC50 = 327.0 μM) and HXH8r (IC50 = 15.32 μM), while exhibiting a remarkable 17.48-fold selectivity for α-glucosidase over α-amylase. Kinetic studies revealed that compound 4d9 acts as a non-competitive inhibitor, and its binding interactions were further investigated using molecular docking analysis. Additionally, compound 4d9 showed noncytotoxic effects on human normal hepatocyte (LO2) cells and demonstrated improved metabolic stability in rat plasma. These findings position compound 4d9 as a promising candidate for the development of therapeutics targeting type 2 diabetes.
Collapse
Affiliation(s)
- Yi-Tong Chen
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Bo-Wen Wan
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kai-Ming Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kong-Kai Zhu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
4
|
Hosseini Nasab N, Raza H, Eom YS, Shah FH, Kwak JH, Kim SJ. Exploring chalcone-sulfonyl piperazine hybrids as anti-diabetes candidates: design, synthesis, biological evaluation, and molecular docking study. Mol Divers 2025; 29:43-59. [PMID: 38775996 DOI: 10.1007/s11030-024-10831-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/25/2024] [Indexed: 02/02/2025]
Abstract
To address the escalating rates of diabetes mellitus worldwide, there is a growing need for novel compounds. The demand for more affordable and efficient methods of managing diabetes is increasing due to the inevitable side effects associated with existing antidiabetic medications. In this present research, various chalcone-sulfonyl piperazine hybrid compounds (5a-k) were designed and synthesized to develop inhibitors against alpha-glucosidase and alpha-amylase. In addition, several spectroscopic methods, including FT-IR, 1H-NMR, 13C-NMR, and HRMS, were employed to confirm the exact structures of the synthesized derivatives. All synthesized compounds were evaluated for their ability to inhibit alpha-glucosidase and alpha-amylase in vitro using acarbose as the reference standard and they showed excellent to good inhibitory potentials. Compound 5k exhibited excellent inhibitory activity against alpha-glucosidase (IC50 = 0.31 ± 0.01 µM) and alpha-amylase (IC50 = 4.51 ± 1.15 µM), which is 27-fold more active against alpha-glucosidase and 7-fold more active against alpha-amylase compared to acarbose, which had IC50 values of 8.62 ± 1.66 µM for alpha-glucosidase and 30.97 ± 2.91 µM for alpha-amylase. It was discovered from the Lineweaver-Burk plot that 5k exhibited competitive inhibition against alpha-glucosidase. Furthermore, cytotoxicity screening assay results against human fibroblast HT1080 cells showed that all compounds had a good level of safety profile. To explore the binding interactions of the most potent compound (5k) with the active site of enzymes, molecular docking research was conducted, and the results obtained supported the experimental data.
Collapse
Affiliation(s)
- Narges Hosseini Nasab
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Hussain Raza
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Young Seok Eom
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Fahad Hassan Shah
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Jae-Hwan Kwak
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea.
| |
Collapse
|
5
|
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
|
6
|
da Silva RB, Borlot JRPO, Rosa Santos C, Rodrigues E Oliveira L, de Almeida LC, Veras Costa-Lotufo L, Octávio Regasini L, Rezende Kitagawa R, de Medeiros EF, de Souza Borges W. New Chalcone Ester Derivatives as Potential Cytotoxic Agents. Chem Biodivers 2024; 21:e202400799. [PMID: 39031575 DOI: 10.1002/cbdv.202400799] [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: 04/01/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/22/2024]
Abstract
Chalcones are a group of molecules with recognized biological potential against many diseases, including cancer. Thus, studies on this structure and derivatives have become an attractive chemical strategy to optimize their observed biological activities. One of the synthetic routes used to obtain chalcone derivatives is esterification using either commercial acid chlorides or carboxylic acids. This work focuses on preparing chalcone derivatives and investigating their biological potential against cancer cells. Compound 3'-hydroxychalcone (1) was synthetized by Claisen-Schmidt condensation followed by esterification of the 3'-OH, resulting in eight compounds named 1a-b and 2a-f. All structures were confirmed by 1H and 13C NMR and FT-IR, and cytotoxicity was evaluated in the HCT 116 (colon adenocarcinoma), MCF-7 (breast adenocarcinoma), and CCD-18Co (nontumoral colon fibroblasts) cell lines. Chalcone derivatives were generally more active toward the colon cancer cell line, and 1a and 2b were selected for IC50 determination, presenting IC50 values of approximately 10 μM in HCT 116 cells and above 20 μM in both MCF7 and CDC-18-Co cells, suggesting moderate selectivity. Additionally, we tested compounds 1a and 2b in combination with doxorubicin, but they did not act synergistically with this anthracycline. In conclusion, considering these compounds obtained by the esterification reaction, 1a and 2d showed better results against cytotoxic cells.
Collapse
Affiliation(s)
- Rafaela Binda da Silva
- Department of Chemistry, Universidade Federal do Espírito Santo (UFES), Avenida Fernando Ferrari 514, 29075-910, Vitória - ES, Brazil
| | | | - Carolina Rosa Santos
- Graduate Program on Chemistry, Universidade Federal do Espírito Santo (UFES), Avenida Fernando Ferrari 514, 29075-910, Vitória - ES, Brazil
| | - Ligia Rodrigues E Oliveira
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, Universidade Estadual Paulista (UNESP), 15054-000, São José do Rio Preto - SP, Brazil
| | - Larissa Costa de Almeida
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), 05508-900, São Paulo - SP, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), 05508-900, São Paulo - SP, Brazil
| | - Luís Octávio Regasini
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, Universidade Estadual Paulista (UNESP), 15054-000, São José do Rio Preto - SP, Brazil
| | - Rodrigo Rezende Kitagawa
- Graduate Program on Chemistry, Universidade Federal do Espírito Santo (UFES), Avenida Fernando Ferrari 514, 29075-910, Vitória - ES, Brazil
| | - Edna Faria de Medeiros
- Department of Chemistry, Universidade Federal do Espírito Santo (UFES), Avenida Fernando Ferrari 514, 29075-910, Vitória - ES, Brazil
| | - Warley de Souza Borges
- Graduate Program on Chemistry, Universidade Federal do Espírito Santo (UFES), Avenida Fernando Ferrari 514, 29075-910, Vitória - ES, Brazil
| |
Collapse
|
7
|
Siddiqui L, Hawsawi MB, Chotana GA, Saleem RSZ. Bis-Chalcones: Recent Reports of Their Diverse Applications in Biological and Material Sciences. ACS OMEGA 2024; 9:42061-42090. [PMID: 39431097 PMCID: PMC11483416 DOI: 10.1021/acsomega.4c04635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024]
Abstract
Originating from the basic chalcone structure, bis-chalcones are characterized by their dual α,β-unsaturated carbonyl systems and carry a range of biological activities that include antimicrobial, antiviral, antiparasitic, antioxidant, antiproliferative, and chemical reactivities that warrant a review to cover recent progress. Thus, this review presents the significant potential demonstrated by bis-chalcones in various biological applications. For example, compounds 2.3.1 showed excellent antiparasitic activity against leishmania with good selectivity index, and compounds 2.2.1-2.2.3 showed submicromolar activity against SupT1 cells. Compound 2.6.22 stood out in its antiproliferative activity against a panel of 60 different cell lines. Compounds 2.6.4 and 2.6.9 have been shown as submicromolar noncompetitive xanthine oxidase inhibitors. We also present their recent applications in material science, for example, as photosensitizers and photoinitiators, to showcase their broader potential for innovation in both medicinal chemistry and industrial applications.
Collapse
Affiliation(s)
- Leena Siddiqui
- Department
of Chemistry and Chemical Engineering, SBASSE, Lahore University of Management Sciences (LUMS), DHA, Lahore-54792, Pakistan
| | - Mohammed B. Hawsawi
- Department
of Chemistry, Faculty of Science, Umm Al-Qura
University, Makkah 21955, Saudi Arabia
| | - Ghayoor Abbas Chotana
- Department
of Chemistry and Chemical Engineering, SBASSE, Lahore University of Management Sciences (LUMS), DHA, Lahore-54792, Pakistan
| | - Rahman Shah Zaib Saleem
- Department
of Chemistry and Chemical Engineering, SBASSE, Lahore University of Management Sciences (LUMS), DHA, Lahore-54792, Pakistan
| |
Collapse
|
8
|
Olender D, Kujawski J, Skóra B, Baranowska-Wójcik E, Sowa-Kasprzak K, Pawełczyk A, Zaprutko L, Szwajgier D, Szychowski KA. Bis-chalcones obtained via one-pot synthesis as the anti-neurodegenerative agents and their effect on the HT-22 cell line. Heliyon 2024; 10:e37147. [PMID: 39286165 PMCID: PMC11403034 DOI: 10.1016/j.heliyon.2024.e37147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
In the area of research on neurodegenerative diseases, the current challenge is to search for appropriate research methods that would detect these diseases at the earliest possible stage, but also new active structures that would reduce the rate of the disease progression and minimize the intensity of their symptoms experienced by the patient. The chalcones are considered in the context of candidates for new drugs dedicated to the fight against neurodegenerative diseases. The synthesis of bis-chalcone derivatives (3a-3d), as aim molecules was performed. Their structures were established by applying 1H NMR, 13C NMR, MS, FT-IR and UV-Vis spectra. All bis-chalcones were synthesized from terephthalaldehyde and appropriate aromatic ketone as substrates in the Claisen-Schmidt condensation method and evaluated in the biological tests and in silico analysis. Compounds exerted antioxidant activity using the HORAC method (3a-3d) and decreased the activities of GPx, COX-2 (3b-3d), GR (3a-3c) and CAT (3a,3b). The high anti-neurodegenerative potential of all four bis-chalcones was observed by inhibition of acetyl- (AChE) and butyrylcholinesterase (BChE) and a positive effect on the mouse hippocampal neuronal HT-22 cell line (LDH release and PGC-1α, PPARγ and GAPDH protein expression). TD-DFT method (computing a number of descriptors associated with HOMO-LUMO electron transition: electronegativity, chemical hardness and potential, first ionization potential, electron affinity) was employed to study the spectroscopic properties. This method showed that the first excited state of compounds was consistent with their maximum absorption in the computed UV-Vis spectra, which showed good agreement with the experimental spectrum using PBE1PBE functional. Using in silico approach, interactions of bis-chalcones with selected targets (aryl hydrocarbon receptor (AhR) PAS-A Domain, ligand binding domain of human PPAR-γ, soman-aged human BChE-butyrylthiocholine complex, Torpedo californica AChE:N-piperidinopropyl-galanthamine complex and the COX-2-celecoxib complex) were characterized. Results obtained in in silico models were consistent with in vitro experiments.
Collapse
Affiliation(s)
- Dorota Olender
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Jacek Kujawski
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszów, Poland
| | - Ewa Baranowska-Wójcik
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8, 20-704, Lublin, Poland
| | - Katarzyna Sowa-Kasprzak
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Anna Pawełczyk
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Lucjusz Zaprutko
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8, 20-704, Lublin, Poland
| | - Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszów, Poland
| |
Collapse
|
9
|
Lv SY, Cheng LP. Design, synthesis and inhibition evaluation of novel chalcone amide α-glucosidase inhibitors. Future Med Chem 2024; 16:1333-1345. [PMID: 39109435 PMCID: PMC11318676 DOI: 10.1080/17568919.2024.2347092] [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: 02/04/2024] [Accepted: 04/09/2024] [Indexed: 08/15/2024] Open
Abstract
Aim: The purpose of this study is to design and synthesize a series of novel chalcone amide α-glucosidase (AG) inhibitors (L1-L10) based on virtual screening and molecular dynamics (MD) simulation. Materials & methods: Target compounds (L1-L10) were synthesized from 2-hydroxyacetophenone and methyl 4-formylbenzoate. Results: In vitro activity test shows that most compounds have good AG inhibition. Specially, compound L4 (IC50 = 8.28 ± 0.04 μM) had the best inhibitory activity, superior to positive control acarbose (IC50 = 8.36 ± 0.02 μM). Molecular docking results show that the good potency of L4 maybe attributed to strong interactions between chalcone skeleton and active site, and the torsion of carbon nitrogen bond in amide group. Conclusion: Compound L4 maybe regard as a good anti-Type II diabetes candidate to preform further study.
Collapse
Affiliation(s)
- Song Yao Lv
- School of Chemical & Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Li Ping Cheng
- School of Chemical & Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China
| |
Collapse
|
10
|
Nian C, Gan X, Liu Q, Wu Y, Kong M, Zhang P, Jin M, Dong Z, Li W, Wang L, He W, Li X, Wu J. Synthesis and Anti-gastric Cancer Activity by Targeting FGFR1 Pathway of Novel Asymmetric Bis-chalcone Compounds. Curr Med Chem 2024; 31:6521-6541. [PMID: 38847254 DOI: 10.2174/0109298673298420240530093525] [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: 12/15/2023] [Revised: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Bis-chalcone compounds with symmetrical structures, either isolated from natural products or chemically synthesized, have multiple pharmacological activities. Asymmetric Bis-chalcone compounds have not been reported before, which might be attributed to the synthetic challenges involved, and it remains unknown whether these compounds possess any potential pharmacological activities. AIMS The aim of this study is to investigate the synthesis route of asymmetric bis-chalcone compounds and identify potential candidates with efficient anti-tumor activity. METHODS The two-step structural optimization of the bis-chalcone compounds was carried out sequentially, guided by the screening of the compounds for their growth inhibitory activity against gastric cancer cells by MTT assay. The QSAR model of compounds was established through random forest (RF) algorithm. The activities of the optimal compound J3 on growth inhibition, apoptosis, and apoptosis-inducing protein expression in gastric cancer cells were investigated sequentially by colony formation assay, flow cytometry, and western blotting. Further, the inhibitory effects of J3 on the FGFR1 signaling pathway were explored by Western Blotting, shRNA, and MTT assays. Finally, the in vivo anti-tumor activity and mechanism of J3 were studied through nude mice xenograft assay, western blotting. RESULTS 27 asymmetric bis-chalcone compounds, including two types (N and J) were sequentially designed and synthesized. Some N-class compounds have good inhibitory activity on the growth of gastric cancer cells. The vast majority of J-class compounds optimized on the basis of N3 exhibit excellent inhibitory activity on gastric cancer cell growth. We established a QSAR model (R2 = 0.851627) by applying random forest algorithms. The optimal compound J3, which had better activity, concentration-dependently inhibited the formation of gastric cancer cell colonies and led to cell apoptosis by inducing the expression of the pro-apoptotic protein cleaved PARP in a dose-dependent manner. J3 may exert anti-gastric cancer effects by inhibiting the activation of FGFR1/ERK pathway. Moreover, at a dose of 10 mg/kg/day, J3 inhibited tumor growth in nude mice by nearly 70% in vivo with no significant toxic effect on body weight and organs. CONCLUSION In summary, this study outlines a viable method for the synthesis of novel asymmetric bischalcone compounds. Furthermore, the compound J3 demonstrates substantial promise as a potential candidate for an anti-tumor drug.
Collapse
Affiliation(s)
- Chunhui Nian
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xin Gan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Department of Pharmacy, Ezhou Central Hospital, Ezhou, Hubei, 436000, China
| | - Qunpeng Liu
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Yuna Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University; Wenzhou, 325027, China
| | - Miaomiao Kong
- The 1st affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiqin Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mingming Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhaojun Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wulan Li
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ledan Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenfei He
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Jianzhang Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University; Wenzhou, 325027, China
| |
Collapse
|
11
|
Nguyen DV, Hengphasatporn K, Danova A, Suroengrit A, Boonyasuppayakorn S, Fujiki R, Shigeta Y, Rungrotmongkol T, Chavasiri W. Structure-yeast α-glucosidase inhibitory activity relationship of 9-O-berberrubine carboxylates. Sci Rep 2023; 13:18865. [PMID: 37914757 PMCID: PMC10620162 DOI: 10.1038/s41598-023-45116-0] [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: 06/24/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023] Open
Abstract
Thirty-five 9-O-berberrubine carboxylate derivatives were synthesized and evaluated for yeast α-glucosidase inhibitory activity. All compounds demonstrated better inhibitory activities than the parent compounds berberine (BBR) and berberrubine (BBRB), and a positive control, acarbose. The structure-activity correlation study indicated that most of the substituents on the benzoate moiety such as methoxy, hydroxy, methylenedioxy, benzyloxy, halogen, trifluoromethyl, nitro and alkyl can contribute to the activities except multi-methoxy, fluoro and cyano. In addition, replacing benzoate with naphthoate, cinnamate, piperate or diphenylacetate also led to an increase in inhibitory activities except with phenyl acetate. 9, 26, 27, 28 and 33 exhibited the most potent α-glucosidase inhibitory activities with the IC50 values in the range of 1.61-2.67 μM. Kinetic study revealed that 9, 26, 28 and 33 interacted with the enzyme via competitive mode. These four compounds were also proved to be not cytotoxic at their IC50 values. The competitive inhibition mechanism of these four compounds against yeast α-glucosidase was investigated using molecular docking and molecular dynamics simulations. The binding free energy calculations suggest that 26 exhibited the strongest binding affinity, and its binding stability is supported by hydrophobic interactions with D68, F157, F158 and F177. Therefore, 9, 26, 28 and 33 would be promising candidates for further studies of antidiabetic activity.
Collapse
Affiliation(s)
- Duy Vu Nguyen
- Department of Chemistry, Faculty of Science, Center of Excellence in Natural Products Chemistry, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Kowit Hengphasatporn
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Ade Danova
- Department of Chemistry, Faculty of Science, Center of Excellence in Natural Products Chemistry, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
- Organic Chemistry Division, Department of Chemistry, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Bandung, West Java, 40132, Indonesia
| | - Aphinya Suroengrit
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Applied Medical Virology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siwaporn Boonyasuppayakorn
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Applied Medical Virology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Ryo Fujiki
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Thanyada Rungrotmongkol
- Bioinformatics and Computational Biology Program, Graduated School, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Biochemistry, Faculty of Science, Center of Excellence in Biocatalyst and Sustainable Biotechnology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Warinthorn Chavasiri
- Department of Chemistry, Faculty of Science, Center of Excellence in Natural Products Chemistry, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
12
|
Kurşun-Aktar BS, Adem Ş, Tatar-Yilmaz G, Hameed ZAH, Oruç-Emre EE. Investigation of α-glucosidase and α-amylase inhibitory effects of phenoxy chalcones and molecular modeling studies. J Mol Recognit 2023; 36:e3061. [PMID: 37720970 DOI: 10.1002/jmr.3061] [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: 06/03/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023]
Abstract
Diabetes mellitus is one of the most critical health problems affecting the quality of life of people worldwide, especially in developing countries. According to the World Health Organization reports, the number of patients with diabetes is approximately 420 million, and this number is estimated to be 642 million in 2040. There are 2 main types of diabetes: Type 1 (T1DM), where the body cannot produce enough insulin, and Type 2 (T2DM), where the body cannot use insulin properly. Patients with T1DM are treated with insulin injections while oral glucose-lowering drugs are used for patients with T2DM. Oral antihyperglycemic drugs used in the treatment of type 2 diabetes mellitus have different mechanisms. Among these, α-Glucosidase and α-amylase inhibitors are one of the most important inhibitors. The antidiabetic effect of the chalcones, which show rich activity, draws attention. This research aims to synthesize chalcone derivatives that could show potential antidiabetic activity. In this study, the inhibitory activity of the chalcone compounds (4a-4g, 5a-5g) was tested against α-glucosidase and α-amylase enzymes. Besides, molecular modeling was utilized to predict potential interactions of the synthesized compounds that exhibit inhibitory effects. In both in vitro and in silico studies, the analyses revealed that compound 5e exhibits strong inhibitory effects against α-glucosidase enzymes (Binding energy: -7.75 kcal/mol, IC50 : 28.88 μM). Additionally, compound 4f demonstrates encouraging inhibitory effects against α-Amylase (Binding energy: -11.08 kcal/mol, IC50 : 46. 21 μM).
Collapse
Affiliation(s)
- Bedriye Seda Kurşun-Aktar
- Department of Hair Care and Beauty Services, Yeşilyurt Vocational School, Malatya Turgut Özal University, Malatya, Turkey
| | - Şevki Adem
- Department of Chemistry, Faculty of Sciences, Çankırı Karatekin University, Çankırı, Turkey
| | - Gizem Tatar-Yilmaz
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | | | - Emine Elçin Oruç-Emre
- Department of Chemistry, Faculty of Art and Sciences, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
13
|
Tran TD, Tu VL, Hoang TM, Dat TV, Tam DNH, Phat NT, Hung DT, Huynh HH, Do TC, Le HH, Minh LHN. A Review of the In Vitro Inhibition of α-Amylase and α-Glucosidase by Chalcone Derivatives. Cureus 2023; 15:e37267. [PMID: 37162770 PMCID: PMC10164439 DOI: 10.7759/cureus.37267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 05/11/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disease relating to steady hyperglycemia resulting from the impairment of the endocrine and non-endocrine systems. Many new drugs having varied targets were discovered to treat this disease, especially type 2 diabetes. Among those, α-glucosidase inhibitors showed their effects by preventing the digestion of carbohydrates through their inhibition against α-amylase and α-glucosidase. Recently, chalcones have attracted considerable attention as they have a simple structure, are easily synthesized as well as have a variety of derivatives. Some reports suggested that chalcone and its derivates could inhibit α-amylase and α-glucosidase. This narrative review provides a comprehensive evaluation of the inhibition of chalcone and its derivatives against α-amylase and α-glucosidase that were reviewed and reported in published scientific articles. Twenty-eight articles were reviewed after screening 207 articles found in four databases, including PubMed, Google Scholar, VHL (Virtual Health Library), and GHL (Global Health Library). This review presented the inhibitory effects of varied chalcones, including chalcones with a basic structural framework, azachalcones, bis-chalcones, chalcone oximes, coumarin-chalcones, cyclohexane chalcones, dihydrochalcones, and flavanone-coupled chalcones. Many of these chalcones had significant inhibition against α-amylase as well as α-glucosidase that were comparable to or even stronger than standard inhibitors. This suggested that such compounds could be potential candidates for the discovery of new anti-diabetic remedies in the years to come.
Collapse
Affiliation(s)
- Thanh-Dao Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Vo Linh Tu
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Thai Minh Hoang
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Truong Van Dat
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Dao Ngoc Hien Tam
- Regulatory Affairs Department, Asia Shine Trading & Service Company Limited, Ho Chi Minh City, VNM
| | - Nguyen Tuan Phat
- Faculty of Medicine, Hue University of Medicine and Pharmacy, Hue, VNM
| | - Dang The Hung
- Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Hong-Han Huynh
- School of Biotechnology, Tan Tao University, Long An, VNM
| | - Thanh C Do
- Faculty of Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, VNM
| | - Huu-Hoai Le
- Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Le Huu Nhat Minh
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, TWN
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei, TWN
| |
Collapse
|
14
|
Pereira R, Silva AMS, Ribeiro D, Silva VLM, Fernandes E. Bis-chalcones: A review of synthetic methodologies and anti-inflammatory effects. Eur J Med Chem 2023; 252:115280. [PMID: 36966653 DOI: 10.1016/j.ejmech.2023.115280] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/03/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Chalcones are bioactive molecules of natural and synthetic sources, whose physicochemical properties, reactivity, and biological activities are well-known among the scientific community. However, there are many molecules strictly related to chalcones with significantly less recognition like bis-chalcones. Several studies indicated that bis-chalcones have advantages over chalcones in specific bioactivities like anti-inflammatory activity. This review article describes the chemical structure and chemical properties of bis-chalcones, as well as the methods reported in the literature for the synthesis of these compounds highlighting the most recent developments. Finally, the anti-inflammatory activity of bis-chalcones is described, emphasizing the active structures found in literature and their mechanisms of action.
Collapse
Affiliation(s)
- Rui Pereira
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Artur M S Silva
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Daniela Ribeiro
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal; Faculty of Agrarian Sciences and Environment, University of the Azores, 9700-042, Angra Do Heroísmo, Açores, Portugal
| | - Vera L M Silva
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Eduarda Fernandes
- LAQV-REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
15
|
Valoy A, Alarcón G, Roco J, Zampini C, Isla MI, Jerez S. A Flavonoid-rich Zuccagnia punctata Extract Prevents High Fat Diet-induced Normal Weight Obesity in a Rabbit Model. PLANTA MEDICA 2023; 89:245-253. [PMID: 35789995 DOI: 10.1055/a-1891-3588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Oral administration of rich in flavonoids hydroalcoholic extract from Zuccagnia punctata (ZpE) improves lipid profile and prevents vascular dysfunction in hypercholesterolemic rabbits. This study aimed to evaluate the ability of ZpE to prevent metabolic and vascular alterations induced by high fat diet (HFD) on a metabolically obese and normal weight rabbit model. The major components of ZpE were analyzed by HPLC method. Rabbits were separated into six groups: 1-fed on standard chow (CD); 2-fed on HFD; 3, 4, 5- fed on HFD and orally administrated 2.5 mg, 5 mg or 10 mg GAE/day of ZpE, respectively (ZpE- HFD); 6- fed on HFD and orally administered 30 mg orlistat/day (Or-HFD). All diets were administrated by 6 weeks. The major compounds of ZpE identified were chalcones: 2',4'-dihydroxy-3'-methoxychalcone and 2',4'-dihydroxychalcone. Oral treatment with ZpE 5 mg GAE/day as well as orlistat prevented the HFD-induced increase of triglycerides, fasting glucose, intraperitoneal glucose test, white cells, and TyG index. Acetylcholine relaxation was reduced in arteries from HFD group and oral administration of ZpE reached this response to CD values. Contractile response to angiotensin II was lower in arteries from rabbits fed on HFD treated with ZpE 5 and 10 mg GAE/day than those of untreated rabbits. Moreover, ZpE could inhibit the activity of pancreatic lipase in vitro and in vivo. In conclusion the ZpE may prevent normal weight obesity by inhibiting the pancreatic lipase. Thus, the use of ZpE as a natural product in the prevention of metabolic syndrome and endothelial dysfunction is very promising.
Collapse
Affiliation(s)
- Agostina Valoy
- Instituto Superior de Investigaciones Biológicas (INSIBIO, UNT-CONICET). San Miguel de Tucumán, Tucumán, Argentina
| | - Gabriela Alarcón
- Instituto Superior de Investigaciones Biológicas (INSIBIO, UNT-CONICET). San Miguel de Tucumán, Tucumán, Argentina
- Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán (UNT). San Miguel de Tucumán, Tucumán, Argentina
| | - Julieta Roco
- Instituto Superior de Investigaciones Biológicas (INSIBIO, UNT-CONICET). San Miguel de Tucumán, Tucumán, Argentina
- Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán (UNT). San Miguel de Tucumán, Tucumán, Argentina
| | - Catiana Zampini
- Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán (UNT). San Miguel de Tucumán, Tucumán, Argentina
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV, UNT-CONICET). San Miguel de Tucumán, Tucumán, Argentina
| | - María Ines Isla
- Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán (UNT). San Miguel de Tucumán, Tucumán, Argentina
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV, UNT-CONICET). San Miguel de Tucumán, Tucumán, Argentina
| | - Susana Jerez
- Instituto Superior de Investigaciones Biológicas (INSIBIO, UNT-CONICET). San Miguel de Tucumán, Tucumán, Argentina
- Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán (UNT). San Miguel de Tucumán, Tucumán, Argentina
| |
Collapse
|
16
|
Moghadam ES, Al-Sadi AM, Al-Harthy T, Faramarzi MA, Shongwe M, Amini M, Abdel-Jalil R. Synthesis, Bioactivity, and Molecular Docking of Benzimidazole-2-carbamate Derivatives as Potent α-Glucosidase Inhibitors. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.134931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
17
|
Wei T, Zheng Z, Wei X, Liu Y, Li W, Fang B, Yun D, Dong Z, Yi B, Li W, Wu X, Chen D, Chen L, Wu J. Rational design, synthesis, and pharmacological characterisation of dicarbonyl curcuminoid analogues with improved stability against lung cancer via ROS and ER stress mediated cell apoptosis and pyroptosis. J Enzyme Inhib Med Chem 2022; 37:2357-2369. [PMID: 36039017 PMCID: PMC9448362 DOI: 10.1080/14756366.2022.2116015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
Curcumin is a natural medicine with a wide range of anti-tumour activities. However, due to β-diketone moiety, curcumin exhibits poor stability and pharmacokinetics which significantly limits its clinical applications. In this article, two types of dicarbonyl curcumin analogues with improved stability were designed through the calculation of molecular stability by density functional theory. Twenty compounds were synthesised, and their anti-tumour activity was screened. A plurality of analogues had significantly stronger activity than curcumin. In particular, compound B2 ((2E,2'E)-3,3'-(1,4-phenylene)bis(1-(2-chlorophenyl)prop-2-en-1-one)) exhibited excellent anti-lung cancer activity in vivo and in vitro. In addition, B2 could upregulate the level of reactive oxygen species in lung cancer cells, which in turn activated the endoplasmic reticulum stress and led to cell apoptosis and pyroptosis. Taken together, curcumin analogue B2 is expected to be a novel candidate for lung cancer treatment with improved chemical and biological characteristics.
Collapse
Affiliation(s)
- Tao Wei
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China
| | - Zhiwei Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Wei
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yugang Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wentao Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bingqing Fang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, China
| | - Di Yun
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhaojun Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Baozhu Yi
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, China
| | - Wulan Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoping Wu
- MOE Key Laboratory of Tumor Molecular Biology, Guangdong, China
| | - Dezhi Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liping Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, China
| | - Jianzhang Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China.,The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Al-Ghulikah HA, Mughal EU, Elkaeed EB, Naeem N, Nazir Y, Alzahrani AYA, Sadiq A, Shah SWA. Discovery of Chalcone Derivatives as Potential α-Glucosidase and Cholinesterase Inhibitors: Effect of Hyperglycemia in Paving a Path to Dementia. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Pérez-González A, Castañeda-Arriaga R, Guzmán-López EG, Hernández-Ayala LF, Galano A. Chalcone Derivatives with a High Potential as Multifunctional Antioxidant Neuroprotectors. ACS OMEGA 2022; 7:38254-38268. [PMID: 36340167 PMCID: PMC9631883 DOI: 10.1021/acsomega.2c05518] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/30/2022] [Indexed: 05/28/2023]
Abstract
A systematic, rational search for chalcone derivatives with multifunctional behavior has been carried out, with the support of a computer-assisted protocol (CADMA-Chem). A total of 568 derivatives were constructed by incorporating functional groups into the chalcone structure. Selection scores were calculated from ADME properties, toxicity, and manufacturability descriptors. They were used to select a subset of molecules (23) with the best drug-like behavior. Reactivity indices were calculated for this subset. They were chosen to account for electron and hydrogen atom donating capabilities, which are key processes for antioxidant activity. The indexes showed that four chalcone derivatives (dCHA-279, dCHA-568, dCHA-553, and dCHA-283) are better electron and H donors than the parent molecule and some reference antioxidants (Trolox, ascorbic acid, and α-tocopherol). In addition, based on molecular docking, they are predicted to act as catechol-O-methyltransferase (COMT), acetylcholinesterase (AChE), and monoamine oxidase B (MAO-B) inhibitors. Therefore, these four molecules are proposed as promising candidates to act as multifunctional antioxidants with neuroprotective effects.
Collapse
Affiliation(s)
- Adriana Pérez-González
- CONACYT
- Universidad Autónoma Metropolitana - Iztapalapa Avenida Ferrocarril
San Rafael Atlixco, número 186, Colonia Leyes de Reforma 1A Sección, Alcaldía Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Romina Castañeda-Arriaga
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Eduardo Gabriel Guzmán-López
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Luis Felipe Hernández-Ayala
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Annia Galano
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| |
Collapse
|
20
|
Rajendran G, Bhanu D, Aruchamy B, Ramani P, Pandurangan N, Bobba KN, Oh EJ, Chung HY, Gangadaran P, Ahn BC. Chalcone: A Promising Bioactive Scaffold in Medicinal Chemistry. Pharmaceuticals (Basel) 2022; 15:1250. [PMID: 36297362 PMCID: PMC9607481 DOI: 10.3390/ph15101250] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chalcones are a class of privileged scaffolds with high medicinal significance due to the presence of an α,β-unsaturated ketone functionality. Numerous functional modifications of chalcones have been reported, along with their pharmacological behavior. The present review aims to summarize the structures from natural sources, synthesis methods, biological characteristics against infectious and non-infectious diseases, and uses of chalcones over the past decade, and their structure-activity relationship studies are detailed in depth. This critical review provides guidelines for the future design and synthesis of various chalcones. In addition, this could be highly supportive for medicinal chemists to develop more promising candidates for various infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Gayathri Rajendran
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Deepu Bhanu
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Baladhandapani Aruchamy
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Prasanna Ramani
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Nanjan Pandurangan
- Department of Sciences, Amrita School of Arts and Sciences, Mysuru Campus, Amrita Vishwa Vidyapeetham, Mysuru 570026, India
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California (San Francisco), San Francisco, CA 94143, USA
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| |
Collapse
|
21
|
Synthesis of Benzofuran–based Schiff bases as anti-diabetic compounds and their molecular docking studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Moghadam ES, Tehrani MH, Abdel-Jalil R, Faramarzi MA, Amini M. Design, Synthesis and Bioactivity Investigation of Novel 2,3-Diarylthiazolidine-4-Ones as Potent α-Glucosidase Inhibitors. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2021.1962369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ebrahim Saeedian Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Maryam Hosseinpour Tehrani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Raid Abdel-Jalil
- Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Xanthone sulfonamide derivatives-A novel series of α-glucosidase inhibitors with different inhibitory types. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
24
|
Mastachi-Loza S, Ramírez-Candelero TI, Benítez-Puebla LJ, Fuentes-Benítes A, González-Romero C, Vázquez MA. Chalcones, a Privileged Scaffold: Highly Versatile Molecules in [4+2] Cycloadditions. Chem Asian J 2022; 17:e202200706. [PMID: 35976743 DOI: 10.1002/asia.202200706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/14/2022] [Indexed: 11/09/2022]
Abstract
Chalcones are aromatic ketones found in nature as the central core of many biological compounds. They have a wide range of biological activity and are biogenetic precursors of other important molecules such as flavonoids. Their pharmacological relevance makes them a privileged scaffold, advantageous for seeking alternative therapies in medicinal chemistry. Due to their structural diversity and ease of synthesis, they are often employed as building blocks for chemical transformations. Chalcones have a carbonyl conjugated system with two electrophilic centers that are commonly used for nucleophilic additions, as described in numerous articles. They can also participate in Diels-Alder reactions, which are [4+2] cycloadditions between a diene and a dienophile. This microreview presents a chronological survey of studies on chalcones as dienes and dienophiles in Diels-Alder cycloadditions. Although these reactions occur in nature, isolation of chalcones from plants yields very small quantities. Contrarily, synthesis leads to large quantities at a low cost. Hence, novel methodologies have been developed for [4+2] cycloadditions, with chalcones serving as a 2π or 4π electron system.
Collapse
Affiliation(s)
- Salvador Mastachi-Loza
- Universidad de Guanajuato Division de Ciencias Naturales y Exactas, Departamento de Química, MEXICO
| | - Tania I Ramírez-Candelero
- Universidad Autonoma del Estado de Mexico Facultad de Quimica, Departamento de Química Orgánica, MEXICO
| | - Luis J Benítez-Puebla
- Universidad de Guanajuato Division de Ciencias Naturales y Exactas, Departamento de Química, MEXICO
| | - Aydee Fuentes-Benítes
- Universidad Autonoma del Estado de Mexico Facultad de Quimica, Departamento de Química Orgánica, MEXICO
| | - Carlos González-Romero
- Universidad Autonoma del Estado de Mexico Facultad de Quimica, Departamento de Química Orgánica, MEXICO
| | - Miguel A Vázquez
- Universidad de Guanajuato Division de Ciencias Naturales y Exactas, CHEMISTRY, NORIA ALTA S/N, 36050, GUANAJUATO, MEXICO
| |
Collapse
|
25
|
Jiang B, Han F, Lü MH, Wang ZP, Liu W, Zhang YX, Xu J, Li RJ. Bis-chalcone polyphenols with potential preventive and therapeutic effects on PD: Design, synthesis and in vitro disaggregation activity against α-synuclein oligomers and fibrils. Eur J Med Chem 2022; 239:114529. [PMID: 35728509 DOI: 10.1016/j.ejmech.2022.114529] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/15/2022]
Abstract
α-Syn fibrils, which are neurotoxic, play a key role in the development of PD. Maintaining α-Syn proteostasis by suitable molecule ligands is an effective approach to prevent aggregation. Disintegrating the existed oligomers and fibrils into individual α-Syn by small molecular compounds is a more efficient way to treat PD. This work designed and synthesized two series of bis-chalcone polyphenol compounds, which possess a sheet-like conjugated skeleton with stronger H-bonding, π-stacking, and hydrophobic interaction with α-Syn protein residues. Some compounds have shown high α-Syn aggregation inhibitory activities in vitro with IC50 down to 0.64 μM. The inhibition goes throughout the aggregation process from the lag to the stationary phase by stabilizing α-Syn proteostasis conformation and preventing β-sheets aggregation, especially in the lag phase. In addition, the inhibitors present good disintegration abilities against the existed α-Syn oligomers and fibrils. The preliminary mechanism studies suggest that the inhibitors could quickly and randomly bind to the specific site closed to the β-sheet domain in the fibril, resulting in unstable and collapse of the protein fibril and yielding a complex system with aggregates of different sizes and monomers. The inhibitors, which could penetrate the blood-brain barrier, are expected to develop into the drug candidates for PD targeting α-Syn aggregation.
Collapse
Affiliation(s)
- Bing Jiang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China
| | - Feng Han
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China
| | - Ming-Huan Lü
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China
| | - Zhen-Ping Wang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China
| | - Wei Liu
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China
| | - Yun-Xiao Zhang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China.
| | - Ji Xu
- Department of Pharmacology, School of Basic Medical Science, Zhengzhou University, Kexue Road 100, 450001, Zhengzhou, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Kexue Road 100, 450001, Zhengzhou, China.
| | - Rui-Jun Li
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Daxue Road 75, 450052, Zhengzhou, China.
| |
Collapse
|
26
|
Simone MI, Wood A, Campkin D, Kiefel MJ, Houston TA. Recent results from non-basic glycosidase inhibitors: How structural diversity can inform general strategies for improving inhibition potency. Eur J Med Chem 2022; 235:114282. [DOI: 10.1016/j.ejmech.2022.114282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/01/2023]
|
27
|
Sohrabi M, Binaeizadeh MR, Iraji A, Larijani B, Saeedi M, Mahdavi M. A review on α-glucosidase inhibitory activity of first row transition metal complexes: a futuristic strategy for treatment of type 2 diabetes. RSC Adv 2022; 12:12011-12052. [PMID: 35481063 PMCID: PMC9020348 DOI: 10.1039/d2ra00067a] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by high blood glucose levels and has emerged as a controversial public health issue worldwide. The increasing number of patients with T2DM on one hand, and serious long-term complications of the disease such as obesity, neuropathy, and vascular disorders on the other hand, have induced a huge economic impact on society globally. In this regard, inhibition of α-glucosidase, the enzyme responsible for the hydrolysis of carbohydrates in the body has been the main therapeutic approach to the treatment of T2DM. As α-glucosidase inhibitors (α-GIs) have occupied a special position in the current research and prescription drugs are generally α-GIs, researchers have been encouraged to design and synthesize novel and efficient inhibitors. Previously, the presence of a sugar moiety seemed to be crucial for designing α-GIs since they can attach to the carbohydrate binding site of the enzyme mimicking the structure of disaccharides or oligosaccharides. However, inhibitors lacking glycosyl structures have also shown potent inhibitory activity and development of non-sugar based inhibitors is accelerating. In this respect, in vitro anti-α-glucosidase activity of metal complexes has attracted lots of attention and this paper has reviewed the inhibitory activity of first-row transition metal complexes toward α-glucosidase and discussed their probable mechanisms of action.
Collapse
Affiliation(s)
- Marzieh Sohrabi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| | | | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences Shiraz Iran
- Central Research Laboratory, Shiraz University of Medical Sciences Shiraz Iran
- Liosa Pharmed Parseh Company Shiraz Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences Tehran Iran
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
28
|
Farwa U, Raza MA. Heterocyclic compounds as a magic bullet for diabetes mellitus: a review. RSC Adv 2022; 12:22951-22973. [PMID: 36105949 PMCID: PMC9379558 DOI: 10.1039/d2ra02697j] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022] Open
Abstract
Diabetes mellitus (DM) is a major metabolic disorder due to hyperglycemia, which is increasing all over the world. From the last two decades, the use of synthetic agents has risen due to their major involvement in curing of chronic diseases including DM. The core skeleton of drugs has been studied such as thiazolidinone, azole, chalcone, pyrrole and pyrimidine along with their derivatives. Diabetics assays have been performed in consideration of different enzymes such as α-glycosidase, α-amylase, and α-galactosidase against acarbose standard drug. The studied moieties were depicted in both models: in vivo as well as in vitro. Molecular docking of the studied compounds as antidiabetic molecules was performed with the help of Auto Dock and molecular operating environment (MOE) software. Amino acid residues Asp349, Arg312, Arg439, Asn241, Val303, Glu304, Phe158, His103, Lys422 and Thr207 that are present on the active sites of diabetic related enzymes showed interactions with ligand molecules. In this review data were organized for the synthesis of heterocyclic compounds through various routes along with their antidiabetic potential, and further studies such as pharmacokinetic and toxicology studies should be executed before going for clinical trials. Diabetes mellitus (DM) is a major metabolic disorder due to hyperglycemia, which is increasing all over the world.![]()
Collapse
Affiliation(s)
- Umme Farwa
- Department of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | | |
Collapse
|
29
|
Tiwari A, Bendi A, Bhathiwal AS. An Overview on Synthesis and Biological Activity of Chalcone Derived Pyrazolines. ChemistrySelect 2021. [DOI: 10.1002/slct.202103779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Aditi Tiwari
- Department of Chemistry Faculty of Science Shree Guru Gobind Singh Tricentenary University Gurugram 122505 Haryana India
| | - Anjaneyulu Bendi
- Department of Chemistry Faculty of Science Shree Guru Gobind Singh Tricentenary University Gurugram 122505 Haryana India
| | - Anirudh Singh Bhathiwal
- Department of Chemistry Faculty of Science Shree Guru Gobind Singh Tricentenary University Gurugram 122505 Haryana India
| |
Collapse
|
30
|
Zhang JH, Xie HX, Li Y, Wang KM, Song Z, Zhu KK, Fang L, Zhang J, Jiang CS. Design, synthesis and biological evaluation of novel (E)-2-benzylidene-N-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl)hydrazine-1-carboxamide derivatives as α-glucosidase inhibitors. Bioorg Med Chem Lett 2021; 52:128413. [PMID: 34634473 DOI: 10.1016/j.bmcl.2021.128413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/25/2021] [Accepted: 10/05/2021] [Indexed: 11/15/2022]
Abstract
In this present study, a series of novel (E)-2-benzylidene-N-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl)hydrazine-1-carboxamide derivatives against α-glucosidase were designed and synthesized, and their biological activities were evaluated in vitro and in vivo. Most of the designed analogues exhibited better inhibitory activity than the marketed acarbose, especially the most potent compound 7 with an IC50 value of 9.26 ± 1.84 μM. The direct binding of 7 and 8 with α-glucosidase was confirmed by fluorescence quenching experiments, and the kinetic and molecular docking studies revealed that 7 and 8 inhibited α-glucosidase in a non-competitive manner. Cytotoxicity bioassay indicated compounds 7 and 8 were non-toxic towards LO2 and HepG2 at 100 μM. Furthermore, both compounds were demonstrated to have in vivo hypoglycemic activity by reducing the blood glucose levels in sucrose-treated rats.
Collapse
Affiliation(s)
- Jin-He Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Hong-Xu Xie
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yue Li
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kai-Ming Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Zhiling Song
- College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Kong-Kai Zhu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Lei Fang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
31
|
Taha M, Alrashedy AS, Almandil NB, Iqbal N, Anouar EH, Nawaz M, Uddin N, Chigurupati S, Wadood A, Rahim F, Das S, Venugopal V, Nawaz F, Khan KM. Synthesis of indole derivatives as diabetics II inhibitors and enzymatic kinetics study of α-glucosidase and α-amylase along with their in-silico study. Int J Biol Macromol 2021; 190:301-318. [PMID: 34481854 DOI: 10.1016/j.ijbiomac.2021.08.207] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/27/2022]
Abstract
In this study, we have investigated a series of indole-based compounds for their inhibitory study against pancreatic α-amylase and intestinal α-glucosidase activity. Inhibitors of carbohydrate degrading enzymes appear to have an essential role as antidiabetic drugs. All analogous exhibited good to moderate α-amylase (IC50 = 3.80 to 47.50 μM), and α-glucosidase inhibitory interactions (IC50 = 3.10-52.20 μM) in comparison with standard acarbose (IC50 = 12.28 μM and 11.29 μM). The analogues 4, 11, 12, 15, 14 and 17 had good activity potential both for enzymes inhibitory interactions. Structure activity relationships were deliberated to propose the influence of substituents on the inhibitory potential of analogues. Docking studies revealed the interaction of more potential analogues and enzyme active site. Further, we studied their kinetic study of most active compounds showed that compounds 15, 14, 12, 17 and 11 are competitive for α-amylase and non- competitive for α-glucosidase.
Collapse
Affiliation(s)
- 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.
| | - Ahlam Sayer Alrashedy
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Noor Barak Almandil
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Naveed Iqbal
- Department of Chemistry, University of Poonch, Rawalakot, AJK, Pakistan
| | - El Hassane Anouar
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Nizam Uddin
- Department of Chemistry, 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, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan
| | - Suprava Das
- Department of Pharmacology, Faculty of Medicine, AIMST University, Semeling, 08100 Bedong, Kedah, Malaysia
| | - Vijayan Venugopal
- School of Pharmacy, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607402, India
| | - Faisal Nawaz
- Department of Chemistry, University of Wah, Quaid Avenue, Wah Cantt 47000, Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
32
|
Saeedian Moghadam E, Sameem B, Abdel-Jalil R, Faramarzi MA, Amini M. 5-Benzylidene-2,3-diarylthiazolidine-4-ones: Design, synthesis, spectroscopic characterization, in vitro biological and computational evaluation. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1946699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ebrahim Saeedian Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Bilqees Sameem
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Raid Abdel-Jalil
- Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Mohammad Ali Faramarzi
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Aroua LM, Almuhaylan HR, Alminderej FM, Messaoudi S, Chigurupati S, Al-Mahmoud S, Mohammed HA. A facile approach synthesis of benzoylaryl benzimidazole as potential α-amylase and α-glucosidase inhibitor with antioxidant activity. Bioorg Chem 2021; 114:105073. [PMID: 34153810 DOI: 10.1016/j.bioorg.2021.105073] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/25/2021] [Accepted: 06/06/2021] [Indexed: 12/23/2022]
Abstract
Synthetic routes to a series of benzoylarylbenzimidazol 3a-h have been derived from 3,4-diaminobenzophenone and an appropriate arylaldehyde in the presence of ammonium chloride or a mixture of ammonium chloride and sodium metabisulfite as catalyst. The antioxidant activity of targeted compounds 3a-h has been measured by four different methods and the overall antioxidant evaluation of the compounds indicated the significant MCA, FRAP, and (DPPH-SA) of the compounds except for the compound 3h. In vitro antidiabetic assay of α-amylase and α-glucosidase suggest a good to excellent activity for most tested compounds. The target benzimidazole 3f containing hydroxyl motif at para-position of phenyl revealed an important activity inhibitor against α- amylase (IC50 = 12.09 ± 0.38 µM) and α-glucosidase (IC50 = 11.02 ± 0.04 µM) comparable to the reference drug acarbose. The results of the anti hyperglycemic activity were supported by means of in silico molecular docking calculations showing strong binding affinity of compounds 3a-h with human pancreatic α-amylase (HPA) and human lysosomal acid-α-glucosidase (HLAG) active sites that confirm a good to excellent activity for most of tested compounds.
Collapse
Affiliation(s)
- Lotfi M Aroua
- Department of Chemistry, College of Science, Qassim University, Campus University, King Abdulaziz Road, Al-Malida, 51452-P.O. Box: 6644, Buraydah, Qassim, Saudi Arabia; Laboratory of Organic Structural Chemistry and Macromolecules, Department of Chemistry, Faculty of Sciences of Tunis, Tunis El-Manar University, El Manar I 2092, Tunis, Tunisia; Carthage University, Faculty of Sciences of Bizerte, 7021 Jarzouna, Tunisia.
| | - Hind R Almuhaylan
- Department of Chemistry, College of Science, Qassim University, Campus University, King Abdulaziz Road, Al-Malida, 51452-P.O. Box: 6644, Buraydah, Qassim, Saudi Arabia
| | - Fahad M Alminderej
- Department of Chemistry, College of Science, Qassim University, Campus University, King Abdulaziz Road, Al-Malida, 51452-P.O. Box: 6644, Buraydah, Qassim, Saudi Arabia
| | - Sabri Messaoudi
- Department of Chemistry, College of Science, Qassim University, Campus University, King Abdulaziz Road, Al-Malida, 51452-P.O. Box: 6644, Buraydah, Qassim, Saudi Arabia; Carthage University, Faculty of Sciences of Bizerte, 7021 Jarzouna, Tunisia
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - Suliman Al-Mahmoud
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
34
|
Nawaz M, Taha M, Qureshi F, Ullah N, Selvaraj M, Shahzad S, Chigurupati S, Abubshait SA, Ahmad T, Chinnam S, Hisaindee S. Synthesis, α-amylase and α-glucosidase inhibition and molecular docking studies of indazole derivatives. J Biomol Struct Dyn 2021; 40:10730-10740. [PMID: 34463216 DOI: 10.1080/07391102.2021.1947892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
Herein, we report the synthesis and inhibitory potential of indazole (Methyl 1H-indazole-4-carboxylate) derivatives (1-13) against α-amylase and α-glucosidase enzymes. The described derivatives demonstrated good inhibitory potential with IC50 values, ranging between 15.04 ± 0.05 to 76.70 ± 0.06 µM ± SEM for α-amylase and 16.99 ± 0.19 to 77.97 ± 0.19 µM ± SEM for α-glucosidase, respectively. In particular, compounds (8-10 and 12) displayed significant inhibitory activities against both the screened enzymes, with their inhibitory potential comparable to the standard acarbose (12.98 ± 0.03 and 12.79 ± 0.17 µM ± SEM, respectively). Additionally, the influence of different substituents on enzyme inhibition activities was assessed to study the structure activity relationships. Molecular docking simulations were performed to rationalize the binding of derivatives/compounds with enzymes. All the synthesized derivatives (1-13) were characterized with the aid of spectroscopic instruments such as 1H-NMR, 13C-NMR, HR-MS, elemental analysis and FTIR.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Faiza Qureshi
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Deanship of Scientific Research, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nisar Ullah
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, Saudi Arabia
| | - Manikandan Selvaraj
- School of Chemical Engineering, Monash University, Selangor Darul Ehsan, Malaysia
| | - Sumaira Shahzad
- School of Business Administration, College of International Education, Zhejiang Gongshang University, Hangzhou, China
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Samar A Abubshait
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Department of Chemistry, College of Science and Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Tauqir Ahmad
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, Saudi Arabia
| | - Sampath Chinnam
- Department of Chemistry, B.M.S. College of Engineering, Bengaluru, Karnataka, India
| | - Soleiman Hisaindee
- Chemistry Department, College of Science, United Arab Emirates University, Al-Ain, UAE
| |
Collapse
|
35
|
Inhibitory Activity and Mechanism Investigation of Hypericin as a Novel α-Glucosidase Inhibitor. Molecules 2021; 26:molecules26154566. [PMID: 34361714 PMCID: PMC8348433 DOI: 10.3390/molecules26154566] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 12/15/2022] Open
Abstract
α-glucosidase is a major enzyme that is involved in starch digestion and type 2 diabetes mellitus. In this study, the inhibition of hypericin by α-glucosidase and its mechanism were firstly investigated using enzyme kinetics analysis, real-time interaction analysis between hypericin and α-glucosidase by surface plasmon resonance (SPR), and molecular docking simulation. The results showed that hypericin was a high potential reversible and competitive α-glucosidase inhibitor, with a maximum half inhibitory concentration (IC50) of 4.66 ± 0.27 mg/L. The binding affinities of hypericin with α-glucosidase were assessed using an SPR detection system, which indicated that these were strong and fast, with balances dissociation constant (KD) values of 6.56 × 10−5 M and exhibited a slow dissociation reaction. Analysis by molecular docking further revealed that hydrophobic forces are generated by interactions between hypericin and amino acid residues Arg-315 and Tyr-316. In addition, hydrogen bonding occurred between hypericin and α-glucosidase amino acid residues Lys-156, Ser-157, Gly-160, Ser-240, His-280, Asp-242, and Asp-307. The structure and micro-environment of α-glucosidase enzymes were altered, which led to a decrease in α-glucosidase activity. This research identified that hypericin, an anthracene ketone compound, could be a novel α-glucosidase inhibitor and further applied to the development of potential anti-diabetic drugs.
Collapse
|
36
|
Bila NM, Costa-Orlandi CB, Vaso CO, Bonatti JLC, de Assis LR, Regasini LO, Fontana CR, Fusco-Almeida AM, Mendes-Giannini MJS. 2-Hydroxychalcone as a Potent Compound and Photosensitizer Against Dermatophyte Biofilms. Front Cell Infect Microbiol 2021; 11:679470. [PMID: 34055673 PMCID: PMC8155603 DOI: 10.3389/fcimb.2021.679470] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
Dermatophytes, fungi that cause dermatophytosis, can invade keratinized tissues in humans and animals. The biofilm-forming ability of these fungi was described recently, and it may be correlated with the long treatment period and common recurrences of this mycosis. In this study, we evaluated the anti-dermatophytic and anti-biofilm activity of 2-hydroxychalcone (2-chalcone) in the dark and photodynamic therapy (PDT)-mediated and to determine its mechanism of action. Trichophyton rubrum and Trichophyton mentagrophytes strains were used in the study. The antifungal susceptibility test of planktonic cells, early-stage biofilms, and mature biofilms were performed using colorimetric methods. Topographies were visualized by scanning electron microscopy (SEM). Human skin keratinocyte (HaCat) monolayers were also used in the cytotoxicity assays. The mechanisms of action of 2-chalcone in the dark and under photoexcitation were investigated using confocal microscopy and the quantification of ergosterol, reactive oxygen species (ROS), and death induction by apoptosis/necrosis. All strains, in the planktonic form, were inhibited after treatment with 2-chalcone (minimum inhibitory concentration (MIC) = 7.8-15.6 mg/L), terbinafine (TRB) (MIC = 0.008–0.03 mg/L), and fluconazole (FLZ) (1–512 mg/L). Early-stage biofilm and mature biofilms were inhibited by 2-chalcone at concentrations of 15.6 mg/L and 31.2 mg/L in all tested strains. However, mature biofilms were resistant to all the antifungal drugs tested. When planktonic cells and biofilms (early-stage and mature) were treated with 2-chalcone-mediated PDT, the inhibitory concentrations were reduced by four times (2–7.8 mg/L). SEM images of biofilms treated with 2-chalcone showed cell wall collapse, resulting from a probable extravasation of cytoplasmic content. The toxicity of 2-chalcone in HaCat cells showed higher IC50 values in the dark than under photoexcitation. Further, 2-chalcone targets ergosterol in the cell and promotes the generation of ROS, resulting in cell death by apoptosis and necrosis. Overall, 2-chalcone-mediated PDT is a promising and safe drug candidate against dermatophytes, particularly in anti-biofilm treatment.
Collapse
Affiliation(s)
- Níura Madalena Bila
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil.,Department of Para-Clinic, School of Veterinary, Universidade Eduardo Mondlane (UEM), Maputo, Mozambique
| | - Caroline Barcelos Costa-Orlandi
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Carolina Orlando Vaso
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Jean Lucas Carvalho Bonatti
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Letícia Ribeiro de Assis
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, Universidade Estadual Paulista (UNESP), Sao Jose do Rio Preto, Brazil
| | - Luís Octavio Regasini
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences, Universidade Estadual Paulista (UNESP), Sao Jose do Rio Preto, Brazil
| | - Carla Raquel Fontana
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Ana Marisa Fusco-Almeida
- Department of Clinical Analysis, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | | |
Collapse
|
37
|
Sousa A, Ribeiro D, Fernandes E, Freitas M. The Effect of Chalcones on the Main Sources of Reactive Species Production: Possible Therapeutic Implications in Diabetes Mellitus. Curr Med Chem 2021; 28:1625-1669. [PMID: 32448100 DOI: 10.2174/0929867327666200525010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus (DM) is characterized by hyperglycaemia, resulting from defects in insulin secretion, insulin action or both. There are several factors such as hyperlipidemia and oxidative stress (OS), namely the production of reactive oxygen/nitrogen species (ROS/RNS), that actively contribute to the development and worsening of DM. Chalcones, also termed as benzalacetophenone or benzylidene acetophenone, present a 1,3-diaryl-2-propen-1-one scaffold that has been shown to be highly promising in the development of new antioxidant compounds. Considering the potential interest of antioxidant therapy, the present review scrutinizes the role of the main sources of ROS/RNS production during DM. The modulatory effect of chalcones against nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine oxidase, mitochondrial respiratory chain and nitric oxide synthase, is also thoroughly discussed, establishing, whenever possible, a structure-activity relationship (SAR). From the SAR analysis, it can be stated that the presence of catechol groups, hydroxyl and methoxyl substituents in the chalcones scaffold improves their modulatory activity against the main sources of ROS/RNS production in DM.
Collapse
Affiliation(s)
- Adelaide Sousa
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
38
|
Shareghi-Boroujeni D, Iraji A, Mojtabavi S, Faramarzi MA, Akbarzadeh T, Saeedi M. Synthesis, in vitro evaluation, and molecular docking studies of novel hydrazineylideneindolinone linked to phenoxymethyl-1,2,3-triazole derivatives as potential α-glucosidase inhibitors. Bioorg Chem 2021; 111:104869. [PMID: 33839583 DOI: 10.1016/j.bioorg.2021.104869] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/02/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023]
Abstract
In this work, a novel series of hydrazineylideneindolinone linked to phenoxymethyl-1,2,3-triazole derivatives were designed, synthesized, and evaluated for their anti-α-glucosidase activity due to an urgent need to develop effective anti-diabetic agents. Among tested 15 compounds, 8 derivatives (9a, 9b, 9c, 9d, 9e, 9f, 9h, and 9o) demonstrated superior potency compared to that of positive control, acarbose. Particularly, compound 9d possessed the best anti-α-glucosidase activity with around a 46-fold improvement in the inhibitory activity. Additionally, 9d showed a competitive type of inhibition in the kinetic study and the molecular docking study demonstrated that it well occupied the binding pocket of the catalytic center through desired interactions with residues, correlating to the experimental results.
Collapse
Affiliation(s)
- Diba Shareghi-Boroujeni
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Iraji
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Alomari M, Taha M, Rahim F, Selvaraj M, Iqbal N, Chigurupati S, Hussain S, Uddin N, Almandil NB, Nawaz M, Khalid Farooq R, Khan KM. Synthesis of indole-based-thiadiazole derivatives as a potent inhibitor of α-glucosidase enzyme along with in silico study. Bioorg Chem 2021; 108:104638. [PMID: 33508679 DOI: 10.1016/j.bioorg.2021.104638] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 12/21/2022]
Abstract
A series of nineteen (1-19) indole-based-thiadiazole derivatives were synthesized, characterized by 1HNMR, 13C NMR, MS, and screened for α-glucosidase inhibition. All analogs showed varied α-glucosidase inhibitory potential with IC50 value ranged between 0.95 ± 0.05 to 13.60 ± 0.30 µM, when compared with the standard acarbose (IC50 = 1.70 ± 0.10). Analogs 17, 2, 1, 9, 7, 3, 15, 10, 16, and 14 with IC50 values 0.95 ± 0.05, 1.10 ± 0.10, 1.30 ± 0.10, 1.60 ± 0.10, 2.30 ± 0.10, 2.30 ± 0.10, 2.80 ± 0.10, 4.10 ± 0.20 and 4.80 ± 0.20 µM respectively showed highest α-glucosidase inhibition. All other analogs also exhibit excellent inhibitory potential. Structure activity relationships have been established for all compounds primarily based on substitution pattern on the phenyl ring. Through molecular docking study, binding interactions of the most active compounds were confirmed. We further studied the kinetics study of analogs 1, 2, 9 and 17 and found that they are Non-competitive inhibitors.
Collapse
Affiliation(s)
- Munther Alomari
- Department of Stem Cell Biology, 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.
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21300, Khyber Pakhtunkhwa, Pakistan
| | - Manikandan Selvaraj
- Monash University School of Chemical Engineering, Bandar Sunway, 47500 Selangor, Malaysia
| | - Naveed Iqbal
- Department of Chemistry University of Poonch, Rawalakot, AJK, Pakistan
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - Shafqat Hussain
- Department of Chemistry, University Of Baltistan, Skardu, Kargil-Skardu Road, Hussainabad, Skardu, Gilgit-Baltistan, Pakistan
| | - Nizam Uddin
- Department of Chemistry, University of Karachi, Karachi 75270, Pakistan
| | - Noor Barak Almandil
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Rai Khalid Farooq
- Department of Neuroscience Research, 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
| |
Collapse
|
40
|
Kaushal R, Kaur M. Bio-medical potential of chalcone derivatives and their metal complexes as antidiabetic agents: a review. J COORD CHEM 2021. [DOI: 10.1080/00958972.2021.1875450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Raj Kaushal
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, India
| | - Mandeep Kaur
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, India
| |
Collapse
|
41
|
Propolis from the Monte Region in Argentina: A Potential Phytotherapic and Food Functional Ingredient. Metabolites 2021; 11:metabo11020076. [PMID: 33525321 PMCID: PMC7911552 DOI: 10.3390/metabo11020076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of this review is to provide overall information on Argentine propolis and to shed light on its potential, especially the one from the Monte region so as to support future research in the field. Around 1999, the Argentine propolis began to be chemically and functionally characterized to give it greater added value. Because Argentina has a wide plant biodiversity, it is expected that its propolis will have various botanical origins, and consequently, a different chemical composition. To date, five types have been defined. Based on their functionality, several products have been developed for use in human and veterinary medicine and in animal and human food. Because the Argentine propolis with the greatest potential is that of the Monte eco-region, this review will describe the findings of the last 20 years on this propolis, its botanical source (Zuccagnia punctata Cav.), its chemical composition, and a description of markers of chemical quality (chalcones) and functionality. Propolis can regulate the activity of various pro-inflammatory enzymes and carbohydrate and lipid metabolism enzymes, as well as remove reactive oxygen and nitrogen species. Consequently, it can modulate metabolic syndrome and could be used as a functional ingredient in food. Furthermore, hydroalcoholic extracts can act against human and animal pathogenic bacteria and human yeast, and mycelial pathogenic fungi. The ability to stop the growth of post-harvest pathogenic bacteria and fungi was also demonstrated. For this reason, Argentine propolis are natural products capable of protecting crops and increasing the lifespan of harvested fruit and vegetables. Several reports indicate the potential of Argentine propolis to be used in innovative products to improve health, food preservation, and packaging. However, there is still much to learn about these natural products to make a wholesome use of them.
Collapse
|
42
|
Salehi B, Quispe C, Chamkhi I, El Omari N, Balahbib A, Sharifi-Rad J, Bouyahya A, Akram M, Iqbal M, Docea AO, Caruntu C, Leyva-Gómez G, Dey A, Martorell M, Calina D, López V, Les F. Pharmacological Properties of Chalcones: A Review of Preclinical Including Molecular Mechanisms and Clinical Evidence. Front Pharmacol 2021; 11:592654. [PMID: 33536909 PMCID: PMC7849684 DOI: 10.3389/fphar.2020.592654] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Chalcones are among the leading bioactive flavonoids with a therapeutic potential implicated to an array of bioactivities investigated by a series of preclinical and clinical studies. In this article, different scientific databases were searched to retrieve studies depicting the biological activities of chalcones and their derivatives. This review comprehensively describes preclinical studies on chalcones and their derivatives describing their immense significance as antidiabetic, anticancer, anti-inflammatory, antimicrobial, antioxidant, antiparasitic, psychoactive, and neuroprotective agents. Besides, clinical trials revealed their use in the treatment of chronic venous insufficiency, skin conditions, and cancer. Bioavailability studies on chalcones and derivatives indicate possible hindrance and improvement in relation to its nutraceutical and pharmaceutical applications. Multifaceted and complex underlying mechanisms of chalcone actions demonstrated their ability to modulate a number of cancer cell lines, to inhibit a number of pathological microorganisms and parasites, and to control a number of signaling molecules and cascades related to disease modification. Clinical studies on chalcones revealed general absence of adverse effects besides reducing the clinical signs and symptoms with decent bioavailability. Further studies are needed to elucidate their structure activity, toxicity concerns, cellular basis of mode of action, and interactions with other molecules.
Collapse
Affiliation(s)
- Bahare Salehi
- Medical Ethics and Law Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de La Salud, Universidad Arturo Prat, Iquique, Chile
| | - Imane Chamkhi
- Faculty of Sciences, Mohammed V University of Rabat, Rabat, Morocco.,Laboratory of Plant-Microbe Interactions, AgroBioSciences, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Abdelaali Balahbib
- Laboratory of Zoology and General Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University Rabat, Rabat, Morocco
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University, Faisalabad, Pakistan
| | - Mehwish Iqbal
- Institute of Health Management, Dow University of Health Sciences, Karachi, Pakistan
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Constantin Caruntu
- Department of Physiology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Department of Dermatology, "Prof. N.C. Paulescu" National Institute of Diabetes, Nutrition, and Metabolic Diseases, Bucharest, Romania
| | - Gerardo Leyva-Gómez
- Departamento De Farmacia, Facultad De Química, Universidad Nacional Autónoma De México, Ciudad De México, Mexico
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, Concepción, Chile.,Unidad De Desarrollo Tecnológico, UDT, Universidad De Concepción, Concepción, Chile
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Víctor López
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain.,Instituto Agroalimentario De Aragón-IA2 CITA-Universidad De Zaragoza, Zaragoza, Spain
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain.,Instituto Agroalimentario De Aragón-IA2 CITA-Universidad De Zaragoza, Zaragoza, Spain
| |
Collapse
|
43
|
Saleem F, Kanwal, Khan KM, Chigurupati S, Solangi M, Nemala AR, Mushtaq M, Ul-Haq Z, Taha M, Perveen S. Synthesis of azachalcones, their α-amylase, α-glucosidase inhibitory activities, kinetics, and molecular docking studies. Bioorg Chem 2021; 106:104489. [PMID: 33272713 DOI: 10.1016/j.bioorg.2020.104489] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 09/23/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
Diabetes being a chronic metabolic disorder have attracted the attention of medicinal chemists and biologists. The introduction of new and potential drug candidates for the cure and treatment of diabetes has become a major concern due to its increased prevelance worldwide. In the current study, twenty-seven azachalcone derivatives 3-29 were synthesized and evaluated for their antihyperglycemic activities by inhibiting α-amylase and α-glucosidase enzymes. Five compounds 3 (IC50 = 23.08 ± 0.03 µM), (IC50 = 26.08 ± 0.43 µM), 5 (IC50 = 24.57 ± 0.07 µM), (IC50 = 27.57 ± 0.07 µM), 6 (IC50 = 24.94 ± 0.12 µM), (IC50 = 27.13 ± 0.08 µM), 16 (IC50 = 27.57 ± 0.07 µM), (IC50 = 29.13 ± 0.18 µM), and 28 (IC50 = 26.94 ± 0.12 µM) (IC50 = 27.99 ± 0.09 µM) demonstrated good inhibitory activities against α-amylase and α-glucosidase enzymes, respectively. Acarbose was used as the standard in this study. Structure-activity relationship was established by considering the parent skeleton and different substitutions on aryl ring. The compounds were also subjected for kinetic studies to study their mechanism of action and they showed competitive mode of inhibition against both enzymes. The molecular docking studies have supported the results and showed that these compounds have been involved in various binding interactions within the active site of enzyme.
Collapse
Affiliation(s)
- Faiza Saleem
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Kanwal
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Institute of Marine Biotechnology, Universiti Malaysia Terengannu, 21030 Kuala Terengganu, Terengganu, Malaysia
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Pakistan Academy of Sciences, 3-Constitution Avenue G-5/2, Islamabad, Pakistan; Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia.
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah 52571, Saudi Arabia
| | - Mehwish Solangi
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Appala Raju Nemala
- Department of Pharmaceutical Chemistry, Sultan-Ul-Uloom College of Pharmacy, Hyderabad, Telangana, India
| | - Maria Mushtaq
- 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 31441, Dammam, Saudi Arabia
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Karachi, Shahra-e-Dr. Salimuzzaman Siddiqui, Karachi 75280, Pakistan
| |
Collapse
|
44
|
Nawaz M, Taha M, Qureshi F, Ullah N, Selvaraj M, Shahzad S, Chigurupati S, Waheed A, Almutairi FA. Structural elucidation, molecular docking, α-amylase and α-glucosidase inhibition studies of 5-amino-nicotinic acid derivatives. BMC Chem 2020; 14:43. [PMID: 32685927 PMCID: PMC7362424 DOI: 10.1186/s13065-020-00695-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/07/2020] [Indexed: 01/18/2023] Open
Abstract
In this study, 5-amino-nicotinic acid derivatives (1-13) have been designed and synthesized to evaluate their inhibitory potential against α-amylase and α-glucosidase enzymes. The synthesized compounds (1-13) exhibited promising α-amylase and α-glucosidase activities. IC50 values for α-amylase activity ranged between 12.17 ± 0.14 to 37.33 ± 0.02 µg/mL ± SEM while for α-glucosidase activity the IC50 values were ranged between 12.01 ± 0.09 to 38.01 ± 0.12 µg/mL ± SEM. In particular, compounds 2 and 4-8 demonstrated significant inhibitory activities against α-amylase and α-glucosidase and the inhibitory potential of these compounds was comparable to the standard acarbose (10.98 ± 0.03 and 10.79 ± 0.17 µg/mL ± SEM, respectively). In addition, the impact of substituent on the inhibitory potential of these compounds was assessed to establish structure activity relationships. Studies in molecular simulations were conducted to better comprehend the binding properties of the compounds. All the synthesized compounds were extensively characterized with modern spectroscopic methods including 1H-NMR, 13C-NMR, FTIR, HR-MS and elemental analysis.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441 Saudi Arabia
| | - 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
| | - Faiza Qureshi
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441 Saudi Arabia
- Deanship of Scientific Research, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441 Saudi Arabia
| | - Nisar Ullah
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, 31261 Saudi Arabia
| | - Manikandan Selvaraj
- School of Chemical Engineering, Monash University, Bandar Subway, 47500 Selangor Darul Ehsan, Malaysia
| | - Sumaira Shahzad
- School of Business Administration, College of International Education, Zhejiang Gongshang University, Hangzhou, China
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, 52571 Saudi Arabia
| | - Abdul Waheed
- Chemistry Department, King Fahd University of Petroleum & Minerals, Dhahran, 31261 Saudi Arabia
| | - Fadiah Ammar Almutairi
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, 52571 Saudi Arabia
| |
Collapse
|
45
|
Abdullah MA, Lee YR, Mastuki SN, Leong SW, Wan Ibrahim WN, Mohammad Latif MA, Ramli ANM, Mohd Aluwi MFF, Mohd Faudzi SM, Kim CH. Development of diarylpentadienone analogues as alpha-glucosidase inhibitor: Synthesis, in vitro biological and in vivo toxicity evaluations, and molecular docking analysis. Bioorg Chem 2020; 104:104277. [PMID: 32971414 DOI: 10.1016/j.bioorg.2020.104277] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/31/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022]
Abstract
A series of aminated- (1-9) and sulfonamide-containing diarylpentadienones (10-18) were synthesized, structurally characterized, and evaluated for their in vitro anti-diabetic potential on α-glucosidase and DPP-4 enzymes. It was found that all the new molecules were non-associated PAINS compounds. The sulfonamide-containing series (compounds 10-18) selectively inhibited α-glucosidase over DPP-4, in which compound 18 demonstrated the highest activity with an IC50 value of 5.69 ± 0.5 µM through a competitive inhibition mechanism. Structure-activity relationship (SAR) studies concluded that the introduction of the trifluoromethylbenzene sulfonamide moiety was essential for the suppression of α-glucosidase. The most active compound 18, was then further tested for in vivo toxicities using the zebrafish animal model, with no toxic effects detected in the normal embryonic development, blood vessel formation, and apoptosis of zebrafish. Docking simulation studies were also carried out to better understand the binding interactions of compound 18 towards the homology modeled α -glucosidase and the human lysosomal α -glucosidase enzymes. The overall results suggest that the new sulfonamide-containing diarylpentadienones, compound 18, could be a promising candidate in the search for a new α-glucosidase inhibitor, and can serve as a basis for further studies involving hit-to-lead optimization, in vivo efficacy and safety assessment in an animal model and mechanism of action for the treatment of T2DM patients.
Collapse
Affiliation(s)
- Maryam Aisyah Abdullah
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Yu-Ri Lee
- Department of Biology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea
| | - Siti Nurulhuda Mastuki
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Sze Wei Leong
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Wan Norhamidah Wan Ibrahim
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Muhammad Alif Mohammad Latif
- Department of Chemistry, Centre of Foundation Studies for Agricultural Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Aizi Nor Mazila Ramli
- Faculty of Industrial Sciences & Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak, Gambang, Kuantan, 26300 Pahang, Malaysia
| | - Mohd Fadhlizil Fasihi Mohd Aluwi
- Faculty of Industrial Sciences & Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak, Gambang, Kuantan, 26300 Pahang, Malaysia
| | - Siti Munirah Mohd Faudzi
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea.
| |
Collapse
|
46
|
Siti Halimatul Munawaroh H, Gumilar GG, Nurjanah F, Yuliani G, Aisyah S, Kurnia D, Wulandari AP, Kurniawan I, Ningrum A, Koyande AK, Show PL. In-vitro molecular docking analysis of microalgae extracted phycocyanin as an anti-diabetic candidate. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107666] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Rocha S, Ribeiro D, Fernandes E, Freitas M. A Systematic Review on Anti-diabetic Properties of Chalcones. Curr Med Chem 2020; 27:2257-2321. [PMID: 30277140 DOI: 10.2174/0929867325666181001112226] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/08/2018] [Accepted: 08/21/2018] [Indexed: 01/22/2023]
Abstract
The use of anti-diabetic drugs has been increasing worldwide and the evolution of therapeutics has been enormous. Still, the currently available anti-diabetic drugs do not present the desired efficacy and are generally associated with serious adverse effects. Thus, entirely new interventions, addressing the underlying etiopathogenesis of type 2 diabetes mellitus, are required. Chalcones, secondary metabolites of terrestrial plants and precursors of the flavonoids biosynthesis, have been used for a long time in traditional medicine due to their wide-range of biological activities, from which the anti-diabetic activity stands out. This review systematizes the information found in literature about the anti-diabetic properties of chalcones, in vitro and in vivo. Chalcones are able to exert these properties by acting in different therapeutic targets: Dipeptidyl Peptidase 4 (DPP-4); Glucose Transporter Type 4 (GLUT4), Sodium Glucose Cotransporter 2 (SGLT2), α-amylase, α-glucosidase, Aldose Reductase (ALR), Protein Tyrosine Phosphatase 1B (PTP1B), Peroxisome Proliferator-activated Receptor-gamma (PPARγ) and Adenosine Monophosphate (AMP)-activated Protein Kinase (AMPK). Chalcones are, undoubtedly, promising anti-diabetic agents, and some crucial structural features have already been established. From the Structure-Activity Relationships analysis, it can generally be stated that the presence of hydroxyl, prenyl and geranyl groups in their skeleton improves their activity for the evaluated anti-diabetic targets.
Collapse
Affiliation(s)
- Sonia Rocha
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313 Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313 Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313 Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313 Porto, Portugal
| |
Collapse
|
48
|
Salas AL, Mercado MI, Eugenia Orqueda M, Correa Uriburu FM, García ME, Pérez MJ, Alvarez MDLA, Ponessa GI, Maldonado LM, Zampini IC, Isla MI. Zuccagnia
‐type Propolis from Argentina: A potential functional ingredient in food to pathologies associated to metabolic syndrome and oxidative stress. J Food Sci 2020. [DOI: 10.1111/1750-3841.15323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Ana L. Salas
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
- Facultad de Ciencias Naturales Universidad Nacional de Tucumán San Miguel de Tucumán Tucumán 4000 Argentina
| | - María Inés Mercado
- Instituto de Morfología Vegetal Área Botánica, Fundación Miguel Lillo San Miguel de Tucumán Tucumán 4000 Argentina
| | - Maria Eugenia Orqueda
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
| | - Florencia M. Correa Uriburu
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
- Facultad de Ciencias Naturales Universidad Nacional de Tucumán San Miguel de Tucumán Tucumán 4000 Argentina
- Instituto Nacional de Tecnología Agropecuaria (INTA) Estación Experimental Agropecuaria Famaillá Famaillá Tucumán 4132 Argentina
| | - Maria Elena García
- Instituto de Morfología Vegetal Área Botánica, Fundación Miguel Lillo San Miguel de Tucumán Tucumán 4000 Argentina
| | - María Jorgelina Pérez
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
| | - María de los Angeles Alvarez
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
| | - Graciela I. Ponessa
- Instituto de Morfología Vegetal Área Botánica, Fundación Miguel Lillo San Miguel de Tucumán Tucumán 4000 Argentina
| | - Luis Maldonado Maldonado
- Instituto Nacional de Tecnología Agropecuaria (INTA) Estación Experimental Agropecuaria Famaillá Famaillá Tucumán 4132 Argentina
| | - Iris Catiana Zampini
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
- Facultad de Ciencias Naturales Universidad Nacional de Tucumán San Miguel de Tucumán Tucumán 4000 Argentina
- Instituto de Morfología Vegetal Área Botánica, Fundación Miguel Lillo San Miguel de Tucumán Tucumán 4000 Argentina
| | - María Inés Isla
- Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV) CONICET‐UNT San Miguel de Tucumán Tucumán 4000 Argentina
- Facultad de Ciencias Naturales Universidad Nacional de Tucumán San Miguel de Tucumán Tucumán 4000 Argentina
- Instituto de Morfología Vegetal Área Botánica, Fundación Miguel Lillo San Miguel de Tucumán Tucumán 4000 Argentina
| |
Collapse
|
49
|
Catharin CW, Chaves AR, de Souza PS, Pérez CN. Babassu activated carbon as catalyst for chalcone production by Claisen–Schmidt reaction: kinetic study, mechanism proposal and continuous flow bed reactor. BRAZILIAN JOURNAL OF CHEMICAL ENGINEERING 2020. [DOI: 10.1007/s43153-020-00034-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
50
|
Cai CY, Zhang W, Wang JQ, Lei ZN, Zhang YK, Wang YJ, Gupta P, Tan CP, Wang B, Chen ZS. Biological evaluation of non-basic chalcone CYB-2 as a dual ABCG2/ABCB1 inhibitor. Biochem Pharmacol 2020; 175:113848. [PMID: 32044354 DOI: 10.1016/j.bcp.2020.113848] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 02/05/2020] [Indexed: 01/23/2023]
Abstract
The enhancement of drug efflux caused by ATP-binding cassette (ABC) transporters (including ABCG2 and ABCB1) overexpression is an important factor for multidrug resistance (MDR) in cancers. After testing the reversal activities of 19 chalcone and bis-chalcone derivatives on MDR cancer cell lines, we found that non-basic chalcone CYB-2 exhibited the most potent reversal activities against both ABCG2- and ABCB1-mediated MDR. The mechanistic studies show that this compound can increase the accumulation of anticancer drugs in both ABCG2- and ABCB1-overexpressing cancer cell lines, resulting from the blocked efflux function of the MDR cancer cell lines. This inhibition is due to the barred ABCG2 and ABCB1 ATPase activities rather than altering the expression or localization of ABCG2 or ABCB1 transporters. The previous studies showed that non-basic chalcones were ABCG2-specific inhibitors; however, we found that non-basic chalcone CYB-2 can be developed as an ABCG2/ABCB1 dual inhibitor to overcome MDR in cancers that co-express both ABCG2 and ABCB1. Moreover, non-basic chalcone CYB-2 has synthetic tractability compared to other chalcone-based derivatives.
Collapse
Affiliation(s)
- Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States; MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, 135 Xingang West Road, Guangzhou 510275, PR China
| | - Wei Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States; Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261041, PR China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States
| | - Yun-Kai Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States
| | - Yi-Jun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States
| | - Pranav Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, 135 Xingang West Road, Guangzhou 510275, PR China.
| | - Bo Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, 135 Xingang West Road, Guangzhou 510275, PR China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States.
| |
Collapse
|