1
|
Ogasawara M, Yoshii K, Wada J, Yamamoto Y, Inouye K. Identification of 2'-deoxyguanosine for an α-amylase inhibitor in the extracts of the earthworm Eisenia fetida and characterization of its inhibitory activity against porcine pancreatic α-amylase. Enzyme Microb Technol 2025; 185:110604. [PMID: 39923336 DOI: 10.1016/j.enzmictec.2025.110604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/06/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Inhibitory activity (IA) against porcine pancreatic α-amylase (PPA) has been found in the water extract of Eisenia fetida. IA is recovered mostly in a fraction (U3EE) containing compounds of low molecular mass under 3 kDa. U3EE is treated with 85 % ethanol (EtOH) and the obtained EtOH extract is applied to the solid-phase extraction (SPE) system. An IA fraction named AI is separated from SPE by elution with water, and then a fraction AI* has been obtained by elution with 5 % EtOH. In the previous study, PPA inhibitors of guanine (Gua), guanosine (Guo), and inosine (Ino) have been isolated from AI. In the present study, IA of AI* was examined and a new inhibitor 2'-deoxyguanosine (2'-dGuo) was isolated by RP-HPLC. It was a mixed-type inhibitor showing IC50 of 0.7 ± 0.1 mM and inhibitor constants Ki and Ki' of 2.0 ± 0.5 and 0.9 ± 0.2 mM, respectively, at pH 6 and at 37 °C. PPA inhibitors of U3EE were arranged in the order according to the inhibitory efficiency as Gua> 2'-dGuo> Guo> Ino. This study reports that compounds related to nucleic acids could be PPA inhibitors. These inhibitors as well as U3EE might be suitable for functional foods to prevent obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Masako Ogasawara
- Research and Development Division, Waki Pharmaceutical Co., Ltd., Room 307, Advanced Chemical Technology Center in Kyoto (ACT Kyoto), 105 Jibu-cho, Fushimi-ku, Kyoto 612-8374, Japan
| | - Katsuhiro Yoshii
- Research and Development Division, Waki Pharmaceutical Co., Ltd., Room 307, Advanced Chemical Technology Center in Kyoto (ACT Kyoto), 105 Jibu-cho, Fushimi-ku, Kyoto 612-8374, Japan
| | - Jun Wada
- Kyoto Municipal Institute of Industrial Technology and Culture, 91 Chudoji Awata-cho, Shimogyo-ku, Kyoto 600-8815, Japan
| | - Yoshihiro Yamamoto
- Kyoto Municipal Institute of Industrial Technology and Culture, 91 Chudoji Awata-cho, Shimogyo-ku, Kyoto 600-8815, Japan
| | - Kuniyo Inouye
- Research and Development Division, Waki Pharmaceutical Co., Ltd., Room 307, Advanced Chemical Technology Center in Kyoto (ACT Kyoto), 105 Jibu-cho, Fushimi-ku, Kyoto 612-8374, Japan.
| |
Collapse
|
2
|
Liang Y, Jin Y, Zhou Y, Zhi J, Wang H. Membrane-bound sucrose hydrolase contributes to carbohydrate metabolism in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 180:104288. [PMID: 40020941 DOI: 10.1016/j.ibmb.2025.104288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Insects mainly rely on sucrase to hydrolyze sucrose into glucose and fructose, supplying carbon and energy for growth and development. Although soluble and membrane-associated sucrases have been identified in several insects, the physiological function of the membrane-bound sucrase remains unclear. Here, we performed a comprehensive analysis of the biochemical properties and physiological functions of the membrane-bound sucrase (BmSUH) in Bombyx mori. Immunofluorescence analysis revealed distinct localization patterns of BmSUH and another crucial sucrase, β-fructofuranosidase (BmSUC1) in the midgut. BmSUH was localized to the microvilli of columnar cells, while BmSUC1 was expressed in the cavities of goblet cells. In addition, the N-terminal transmembrane domain is crucial for membrane localization of BmSUH. We then verified that one of the positive selection sites, N326, is N-glycosylated and essential for the enzyme activity of BmSUH. CRISPR/Cas9-mediated knockout of BmSUH significantly reduced both membrane-associated and membrane-bound sucrase activity in the midgut, leading to decreased sucrose absorption from food. Transcriptome analysis further revealed the molecular mechanisms underlying the physiological function of BmSUH, with differentially expressed genes enriched in many pathways related to digestion, absorption, and metabolism of carbohydrates. These results highlight that BmSUH served as an essential sucrase involved in the digestive and metabolic processes. This study provides insight into the functional evolution of the membrane-bound sucrase and advances our understanding of sucrose utilization in lepidopteran insects.
Collapse
Affiliation(s)
- Yanting Liang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue Jin
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanyan Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jinsi Zhi
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Huabing Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
3
|
Dhanya TM, Prathapachandra Kurup MR, Rajimon KJ, Anjali Krishna G, Varughese JK, Raghu KG, Philip S, Divya KM, Augustine M, Mohanan PV. Unveiling the multifaceted bioactivity of copper(II)-Schiff base complexes: a comprehensive study of antioxidant, anti-bacterial, anti-inflammatory, enzyme inhibition and cytotoxic potentials with DFT insights. Dalton Trans 2025; 54:3216-3234. [PMID: 39820950 DOI: 10.1039/d4dt02486a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The rise of various diseases demands the development of new agents with antioxidant, antimicrobial, anti-inflammatory, enzyme-inhibiting, and cytotoxic properties. In this study, heterocyclic Schiff base complexes of Cu(II) featuring a benzo[b]thiophene moiety were synthesized and their biological activities evaluated. The complexes were characterized using FT-IR, UV-Vis, and EPR spectroscopy, TG-DTG analysis, magnetic moment measurements, molar conductivity measurements, and elemental analyses. Density functional theory (DFT) calculations were used to optimize the theoretical molecular orbital energies of the copper complexes. The complexes exhibited square pyramidal and square planar geometries. Biological assays demonstrated that these complexes generally outperformed the Schiff base ligands for various activities. The antioxidant capacity, measured via the DPPH assay in methanol, was comparable to those of the BHT and ascorbic acid standards, with 4BNPC showing the lowest IC50 value, which was attributed to the free OH group rather than coordination to the metal center. The anti-bacterial activity was assessed using the agar disc diffusion method against E. coli, P. aeruginosa, B. subtilis, and S. aureus, with BAC showing the largest inhibition zone compared to the others and ciprofloxacin as the reference. The anti-inflammatory activity, evaluated by the HRBC membrane stabilization method, showed that the 4BNPC Cu(II) complex had moderate activity similar to that of diclofenac. Enzyme inhibition studies against α-amylase revealed that the BAC complexes had the highest inhibition values, surpassing those of the Schiff base ligands. Cytotoxicity was assessed using Trypan blue exclusion for DLA and HepG2 cancer cell lines, and the MTT assay for H9c2 human cells. BMPC demonstrated superior cytotoxicity at both high and low concentrations against the normal H9c2 cell line. Among the tested compounds, BNPC showed moderate inhibition against HepG2 cells, while BMPC exhibited the greatest cytotoxicity at higher concentrations, particularly reaching nearly 100% cell death at 200 μg mL-1 in DLA cell lines. This suggests that BMPC is a promising candidate for further pharmacological research, particularly against DLA cells.
Collapse
Affiliation(s)
- T M Dhanya
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
| | - M R Prathapachandra Kurup
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
| | - K J Rajimon
- Department of Chemistry, St Berchmans College, Changanacherry, Kerala, India.
| | - G Anjali Krishna
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
- Department of Science and Humanities, Mar Baselios Institute of Technology and Science, Nellimattom, Kothamangalam, Kerala, India.
| | | | - K G Raghu
- Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, Kerala, India.
| | - Sachin Philip
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
| | - K M Divya
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
- Department of Chemistry, NSS College, Cherthala, Kerala, India.
| | - Maria Augustine
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
- Department of Chemistry, St Paul's College, Kalamassery, Kerala, India.
| | - P V Mohanan
- Department of Applied Chemistry, Cochin University of Science and Technology, Kochi 22, Kerala, India.
| |
Collapse
|
4
|
Singh R, Sindhu J, Singh D, Kumar P. Key molecular scaffolds in the development of clinically viable α-amylase inhibitors. Future Med Chem 2025; 17:347-362. [PMID: 39835704 PMCID: PMC11792802 DOI: 10.1080/17568919.2025.2453421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
The escalating cases of type II diabetes combined with adverse side effects of current antidiabetic drugs spurred the advancement of innovative approaches for the management of postprandial glucose levels. α-Amylase is an endoamylase responsible for the breakdown of internal α-1,4-glycosidic linkages in dietary starch, producing oligosaccharides. Subsequently, α-glucosidase degraded these oligosaccharides to monosaccharides, which are absorbed into the bloodstream and become available to the body. The inhibitors of α-amylase reduced the digestibility of carbohydrates accompanied by delayed glucose absorption, leading to decreased blood glucose levels after meals and thus, inhibition of the enzyme seems to be a crucial strategy for diabetes management and improving overall glycemic control in diabetic patients. The present review article emphasizes the therapeutic promise of recently discovered potential α-amylase inhibitors, highlighting their in vitro, in silico and in vivo profiles. Ultimately, we addressed the contemporary challenges and potential routes ahead in the search for safe and reliable α-amylase inhibitors for clinical use, summarizing the most recent research in the field.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
- School of Chemistry, Indian Institutes of Science Education and Research, Thiruvananthapuram, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
5
|
Jain S, Sharma MK, Gupta N, Anirudh J, Banavath HN, Chatterjee S. An experimental and computational approach to evaluate the antidiabetic activity of Commiphora wightii gum extract. J Ayurveda Integr Med 2025; 16:101038. [PMID: 39705799 PMCID: PMC11910329 DOI: 10.1016/j.jaim.2024.101038] [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: 11/28/2023] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Plant formulations with antidiabetic and antioxidant properties have recently gained popularity due to their lower cost and lesser side effects. Guggul gum is one such formulation that is extensively being used to cure various ailments. OBJECTIVE The present study was designed to explore the antioxidant and antidiabetic properties of the aqua-ethanolic Guggul gum extract (GE) from Commiphora wightii using in silico studies and in vitro assays. METHODS Gas Chromatography Mass Spectroscopy (GCMS) identified compounds were docked to the Human pancreatic α-amylase (HPA, PDB ID: 1HNY) for in silico studies to predict the inhibition. Molecular dynamics simulations (MDS) were performed using GROMACS for 100 ns. The inhibition of the enzyme was further evaluated at in vitro level to show the compounds' hypoglycemic role. RESULTS The extract showed a good amount of phenolic (5.14 ± 0.011 mg), flavonoid (0.66 ± 0.023 mg) and terpenoid (1.08 ± 0.018 mg) content along with a promising free radical scavenging activity of 41.96 ± 4.02%. In the in silico studies, 3 out of 6 GCMS-identified bioactive compounds showed permissible values of bioavailability properties suggesting them as a potential candidate for antidiabetic drugs. Similarly, in molecular docking studies, 3 compounds showed more binding energy than the standard drug acarbose indicating better inhibition. MDS studies showed Compound 4 (Diisooctyl phthalate), was the most stable with the lowest root mean square deviation (RMSD) and root mean square fluctuation (RMSF) values, a consistent radius of gyration (Rg), and stable solvent accessible surface area (SASA). This was further confirmed by in vitro analysis where the pancreatic α-amylase inhibitory activity of the extract and the standard drug (acarbose) were comparable at an IC50 value of 4.17 ± 1.26 mg/mL and 3.69 ± 0.89 mg/mL respectively. CONCLUSION The results demonstrated GE as a potential alternative to commercial antidiabetic drugs. Out of the major 6 GCMS-identified compounds, Compound 4 showed the most stable conformation during MDS studies. However, the isolation of the identified compounds could be done in the future for in vivo studies.
Collapse
Affiliation(s)
- Shalini Jain
- Department of Zoology, IIS (deemed to be University), Jaipur, 302020, India
| | | | - Nidhi Gupta
- Department of Microbiology & Biotechnology, IIS (deemed to be University), Jaipur, 302020, India
| | - Jivanage Anirudh
- Department of Sports Biosciences, School of Sports Science Yoga & Education, Central University of Rajasthan, Ajmer, 305817, India
| | - Hemanth Naick Banavath
- Department of Sports Biosciences, School of Sports Science Yoga & Education, Central University of Rajasthan, Ajmer, 305817, India
| | - Sreemoyee Chatterjee
- Department of Microbiology & Biotechnology, IIS (deemed to be University), Jaipur, 302020, India.
| |
Collapse
|
6
|
Yadav N, Kumar R, Sangwan S, Dhanda V, Duhan A, Sindhu J. Environment benign synthesis of 5-acyl-4-hydroxypyridin-2(1 H)-one derivatives as antioxidant and α-amylase inhibitors. Future Med Chem 2024; 16:2637-2646. [PMID: 39606936 PMCID: PMC11734593 DOI: 10.1080/17568919.2024.2432289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
AIM Oxidative stress, caused by postprandial activities, is a major global health issue causing chronic diseases like diabetes mellitus, cancer, and asthma. Therefore, it was envisaged to design and synthesize a series of substituted 4-hydroxypyridine-2(1 h)-ones in order to develop new molecules that can reduce oxidative stress and modulate α-amylase activity also. MATERIALS & METHODS An environmentally benign, solvent and catalyst free, natural product inspired synthesis of 4-hydroxypyridin-2(1 h)-one derivatives has been developed. The synthetic analogues were evaluated in vitro α-amylase activity and antioxidant potential. RESULTS Among all the synthesized compounds, 4a, 4c, and 4d displayed many folds higher antioxidants activity than the standard, BHT. The in vitro α-amylase inhibition was found to be moderate with IC50 values ranging from 5.48 to 9.31 mm as compared to the standard acarbose (IC50 = 0.65 mm). The most active compound against α-amylase 4c was further investigated for its binding affinity within the active site of the enzyme and the kinetics studies revealed probable uncompetitive mode of inhibition. CONCLUSION Compound 4a was found to be promising antioxidant and 4c as a good α-amylase inhibitor. These compounds could pave the way for development of new α-amylase inhibitors with antioxidant capabilities thereby effectively mitigating diabetes mellitus.
Collapse
Affiliation(s)
- Neelam Yadav
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
| | - Ravi Kumar
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
- MAP Section, Department of Genetics and Plant Breeding, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
- Center for Bio-Nanotechnology, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
| | - Sarita Sangwan
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
| | - Vidhi Dhanda
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
| | - Anil Duhan
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
| | - Jayant Sindhu
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar, Hisar, Haryana, India
| |
Collapse
|
7
|
Visvanathan R, Le DT, Dhital S, Rali T, Davis RA, Williamson G. Inhibition of Human Salivary and Pancreatic α-Amylase by Resveratrol Oligomers. J Med Chem 2024; 67:18753-18763. [PMID: 39501642 PMCID: PMC11571111 DOI: 10.1021/acs.jmedchem.4c01042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024]
Abstract
A key strategy to mitigate postprandial hyperglycemia involves inhibiting α-amylases, which commence the starch digestion process in the gut. This study examined the inhibitory effects of resveratrol and stilbenoid tetramers, vaticanol B, (-)-hopeaphenol, and vatalbinoside A on human salivary and pancreatic α-amylases experimentally and through molecular docking studies. Vaticanol B demonstrated the most potent inhibition with IC50 values of 5.3 ± 0.3 μM for salivary and 6.1 ± 0.5 μM for pancreatic α-amylase (compared to acarbose with IC50 values of 1.2 ± 0.1 μM and 0.5 ± 0.0 μM, respectively). Kinetic analysis suggested a competitive inhibition mode for vaticanol B. Resveratrol and vatalbinoside A were poor inhibitors of human α-amylases, while (-)-hopeaphenol exhibited moderate inhibition. Molecular docking supported the inhibition data, and several aspects of the structural configurations explained the stronger inhibition exerted by vaticanol B. Overall, vaticanol B shows promise as a natural alternative to acarbose for inhibiting α-amylase.
Collapse
Affiliation(s)
- Rizliya Visvanathan
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
| | - Dang Truong Le
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
- Bioresource
Processing Research Institute of Australia (BioPRIA), Department of
Chemical and Biological Engineering, Monash
University, Clayton, VIC 3800, Australia
| | - Sushil Dhital
- Bioresource
Processing Research Institute of Australia (BioPRIA), Department of
Chemical and Biological Engineering, Monash
University, Clayton, VIC 3800, Australia
| | - Topul Rali
- School
of Natural and Physical Sciences, The University
of Papua New Guinea, Port Moresby, Papua New Guinea
| | - Rohan A. Davis
- Institute
for Biomedicine and Glycomics, Griffith
University, Brisbane, QLD 4111, Australia
| | - Gary Williamson
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
| |
Collapse
|
8
|
Zhang L, Li Z, Kong H, Ban X, Gu Z, Hong Y, Cheng L, Li C. Advances in microbial exopolysaccharides as α-amylase inhibitors: Effects, structure-activity relationships, and anti-diabetic effects in vivo. Int J Biol Macromol 2024; 281:136174. [PMID: 39366595 DOI: 10.1016/j.ijbiomac.2024.136174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/22/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
The rapid digestion of starch, as the main source of energy in the human diet, causes an acute increase in blood sugar levels that will affect blood glucose homeostasis. The inhibition of α-amylase activity is an effective way of reducing starch digestibility, thereby controlling postprandial glycemia. As a class of carbohydrate polymers, microbial exopolysaccharides (EPSs) have garnered widespread attention for their inhibitory effects on α-amylase, but there is a lack of comprehensive review in this area. This paper aimed to review the inhibitory activity of microbial EPSs on α-amylase and their interaction mechanisms, and the effect of microbial EPSs on lowering blood glucose levels and regulating glycolipid metabolism in vivo were also discussed. Numerous studies have reported that EPSs with α-amylase inhibition activity are primarily produced by lactic acid bacteria. Microbial EPSs with an appropriate range of molecular weight, high proportion of glucose or mannose or arabinose residues, and high uronic acid content might be acceptable to inhibit α-amylase activity. Additionally, microbial EPSs exhibited potential anti-diabetic effects in mice, reducing blood glucose levels, and regulating glycolipid metabolism and gut microbiota. The information covered in this review may enhance the development and application of EPSs in functional food and pharmaceutical research.
Collapse
Affiliation(s)
- Lan Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing 214200, China
| | - Haocun Kong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiaofeng Ban
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Caiming Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing 214200, China.
| |
Collapse
|
9
|
Cao J, Zhang J, Cao R, Zhang B, Miao M, Liu X, Sun L. Enzymolysis Modes Trigger Diversity in Inhibitor-α-Amylase Aggregating Behaviors and Activity Inhibition: A New Insight Into Enzyme Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404127. [PMID: 39234852 PMCID: PMC11538681 DOI: 10.1002/advs.202404127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/17/2024] [Indexed: 09/06/2024]
Abstract
Inhibitors of α-amylase have been developed to regulate postprandial blood glucose fluctuation. The enzyme inhibition arises from direct or indirect inhibitor-enzyme interactions, depending on inhibitor structures. However, an ignored factor, substrate, may also influence or even decide the enzyme inhibition. In this work, it is innovatively found that the difference in substrate enzymolysis modes, i.e., structural composition and concentration of α-1,4-glucosidic bonds, triggers the diversity in inhibitor-enzyme aggregating behaviors and α-amylase inhibition. For competitive inhibition, there exists an equilibrium between α-amylase-substrate catalytic affinity and inhibitor-α-amylase binding affinity; therefore, a higher enzymolysis affinity and concentration of α-1,4-glucosidic structures interferes the balance, unfavoring inhibitor-enzyme aggregate formation and thus weakening α-amylase inhibition. For uncompetitive inhibition, the presence of macromolecular starch is necessary instead of micromolecular GalG2CNP, which not only binds with active site but with an assistant flexible loop (involving Gly304-Gly309) near the site. Hence, the refined enzyme structure due to the molecular flexibility more likely favors the inhibitor binding with the non-active loop, forming an inhibitor-enzyme-starch ternary aggregate. Conclusively, this study provides a novel insight into the evaluation of α-amylase inhibition regarding the participating role of substrate in inhibitor-enzyme aggregating interactions, emphasizing the selection of appropriate substrates in the development and screening of α-amylase inhibitors.
Collapse
Affiliation(s)
- Junwei Cao
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingShaanxi712100China
| | - Jifan Zhang
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingShaanxi712100China
| | - Ruibo Cao
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingShaanxi712100China
| | - Bin Zhang
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Ming Miao
- State Key Laboratory of Food Science and ResourcesJiangnan University1800 Lihu AvenueWuxiJiangsu214122China
| | - Xuebo Liu
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingShaanxi712100China
| | - Lijun Sun
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingShaanxi712100China
| |
Collapse
|
10
|
Mayo-Montor CI, Vidal-Limon A, Loyola-Vargas VM, Carmona-Hernández O, Barreda-Castillo JM, Monribot-Villanueva JL, Guerrero-Analco JA. Targeting Hypoglycemic Natural Products from the Cloud Forest Plants Using Chemotaxonomic Computer-Assisted Selection. Int J Mol Sci 2024; 25:10881. [PMID: 39456663 PMCID: PMC11507857 DOI: 10.3390/ijms252010881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The cloud forest (CF), a hugely biodiverse ecosystem, is a hotspot of unexplored plants with potential for discovering pharmacologically active compounds. Without sufficient ethnopharmacological information, developing strategies for rationally selecting plants for experimental studies is crucial. With this goal, a CF metabolites library was created, and a ligand-based virtual screening was conducted to identify molecules with potential hypoglycemic activity. From the most promising botanical families, plants were collected, methanolic extracts were prepared, and hypoglycemic activity was evaluated through in vitro enzyme inhibition assays on α-amylase, α-glucosidase, and dipeptidyl peptidase IV (DPP-IV). Metabolomic analyses were performed to identify the dominant metabolites in the species with the best inhibitory activity profile, and their affinity for the molecular targets was evaluated using ensemble molecular docking. This strategy led to the identification of twelve plants (in four botanical families) with hypoglycemic activity. Sida rhombifolia (Malvaceae) stood out for its DPP-IV selective inhibition versus S. glabra. A comparison of chemical profiles led to the annotation of twenty-seven metabolites over-accumulated in S. rhombifolia compared to S. glabra, among which acanthoside D and cis-tiliroside were noteworthy for their potential selective inhibition due to their specific intermolecular interactions with relevant amino acids of DPP-IV. The workflow used in this study presents a novel targeting strategy for identifying novel bioactive natural sources, which can complement the conventional selection criteria used in Natural Product Chemistry.
Collapse
Affiliation(s)
- Cecilia I. Mayo-Montor
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C., Xalapa 91073, Mexico; (C.I.M.-M.); (A.V.-L.); (J.M.B.-C.)
| | - Abraham Vidal-Limon
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C., Xalapa 91073, Mexico; (C.I.M.-M.); (A.V.-L.); (J.M.B.-C.)
| | | | | | - José Martín Barreda-Castillo
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C., Xalapa 91073, Mexico; (C.I.M.-M.); (A.V.-L.); (J.M.B.-C.)
| | - Juan L. Monribot-Villanueva
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C., Xalapa 91073, Mexico; (C.I.M.-M.); (A.V.-L.); (J.M.B.-C.)
| | - José A. Guerrero-Analco
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C., Xalapa 91073, Mexico; (C.I.M.-M.); (A.V.-L.); (J.M.B.-C.)
| |
Collapse
|
11
|
Kumari S, Saini R, Mishra A. Phytochemical profiling and evaluation of the antidiabetic potential of Ichnocarpus frutescens (Krishna Sariva): kinetic study, molecular modelling, and free energy approach. J Biomol Struct Dyn 2024; 42:8712-8731. [PMID: 37615387 DOI: 10.1080/07391102.2023.2248265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
This research explored novel antidiabetic drugs from natural sources using the Ayurvedic Rasayana herb Ichnocarpus frutescens through invitro enzyme assay, kinetics study, and computational approaches. Invitro enzyme inhibition assay demonstrated the promising inhibitory activity of root extract against alpha-amylase (α-A) and alpha-glucosidase (α-G) enzyme with IC50 value 7.34 ± 0.22 mg/ml and 4.40 ± 0.25 mg/ml respectively. Enzyme kinetic study revealed the competitive inhibition of both proteins by Ichnocarpus frutescens extract. High-Resolution Liquid Chromatography Mass Spectrometer and Docking study revealed the better binding energy of phytoconstituents 23-Acetoxysoladulcidine, Atrovirinone, Bismurrayaquinone A, Lamprolobine, Zygadenine, and Gambiriin A3 than standard drug acarbose. Molecular modelling showed stable protein-ligands binding interaction during the 100 ns simulation. It revealed comparable Root Mean Square Deviation, Radius of Gyration, and Solvent Accessible Surface Area of these compounds with acarbose. The active site residues of both proteins remained stable and showed significantly less Root Mean Square Fluctuation. Molecular Mechanics with Generalised Bonn Surface Area analysis has illustrated the similar inhibitory activity of Zygadenine for α-A, 23-Acetoxysoladulcidine, and Gambiriin A3 for α-G protein, compared to the FDA-approved drug acarbose. Thus, the study suggested that the root of Ichnocarpus frutescens can be used as α-A and α-G inhibitors and be considered a compelling lead for the medication of type 2 diabetes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sonali Kumari
- aSchool of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Ravi Saini
- aSchool of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Abha Mishra
- aSchool of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| |
Collapse
|
12
|
Choi H, Kim S, Park T, Lee SH. Fluorogenic Monitoring of α-Amylase in Human Urine for Straightforward Diagnosis of Pancreatic Diseases. Chem Asian J 2024; 19:e202400505. [PMID: 38959126 DOI: 10.1002/asia.202400505] [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: 04/30/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024]
Abstract
In this study, we developed a sensitive method for monitoring α-amylase using a fluorogenic approach based on the host-guest complexation between an amphiphilic pyrenyl derivative (1) and γ-cyclodextrins (γ-CDs). The compound 1 self-assembles into nanofibrils in aqueous solutions. Upon the introduction of γ-CD, compound 1 forms an inclusion complex with it. This complex then participates in the formation of a 2 : 2 complex with another complex, leading to strong excimer fluorescence. Upon interaction with α-amylase, γ-CD undergoes hydrolysis, leading to the regeneration of nanofibrils, which is accompanied by a decrease in excimer fluorescence and an increase in monomeric fluorescence. This ratiometric fluorescence color change enables the sensitive detection of low levels of α-amylase in human urine, offering a practical approach for early screening of pancreatic-related diseases.
Collapse
Affiliation(s)
- Haemin Choi
- Department of Chemistry, Daegu University, Gyeongsan, 38453, Republic of Korea
| | - Seoyun Kim
- Department of Chemistry, Daegu University, Gyeongsan, 38453, Republic of Korea
| | - Taemin Park
- Department of Chemistry, Daegu University, Gyeongsan, 38453, Republic of Korea
| | - Seoung Ho Lee
- Department of Chemistry, Daegu University, Gyeongsan, 38453, Republic of Korea
| |
Collapse
|
13
|
Gavanji S, Bakhtari A, Abdel-Latif R, Bencurova E, Othman EM. Experimental approaches for induction of diabetes mellitus and assessment of antidiabetic activity: An in vitro and in vivo methodological review. Fundam Clin Pharmacol 2024; 38:842-861. [PMID: 38747157 DOI: 10.1111/fcp.13009] [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: 09/14/2023] [Revised: 02/26/2024] [Accepted: 04/25/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Diabetes mellitus poses a global health challenge, driving the need for innovative therapeutic solutions. Experimental methods play a crucial role in evaluating the efficacy of potential antidiabetic drugs, both in vitro and in vivo. Yet concerns about reproducibility persist, necessitating comprehensive reviews. OBJECTIVES This review aims to outline experimental approaches for inducing diabetes and evaluating antidiabetic activity, synthesizing data from authoritative sources and academic literature. METHODS We conducted a systematic search of prominent databases, including PubMed, ScienceDirect, and Scopus, to identify relevant articles spanning from 1943 to the present. A total of 132 articles were selected for inclusion in this review, focusing on in vitro and in vivo experimental validations of antidiabetic treatments. RESULTS Our review highlights the diverse array of experimental methods employed for inducing diabetes mellitus and evaluating antidiabetic interventions. From cell culture assays to animal models, researchers have employed various techniques to study the effectiveness of novel therapeutic agents. CONCLUSION This review provides a comprehensive guide to experimental approaches for assessing antidiabetic activity. By synthesizing data from a range of sources, we offer valuable insights into the current methodologies used in diabetes research. Standardizing protocols and enhancing reproducibility are critical for advancing effective antidiabetic treatments.
Collapse
Affiliation(s)
- Shahin Gavanji
- Department of Plant Biotechnology, Medicinal Plants Research Centre, Islamic Azad University, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Azizollah Bakhtari
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rania Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Elena Bencurova
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Eman M Othman
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
14
|
Nicol A, Ahmed M, Fischer C, Garces JG, Magnus S, Maung N, Molisani N, Petrov S, Palu RAS. Larval density can be used to predict genetic modifiers of glucagon signaling in Drosophila melanogaster. PLoS One 2024; 19:e0302565. [PMID: 39196987 PMCID: PMC11356449 DOI: 10.1371/journal.pone.0302565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 04/05/2024] [Indexed: 08/30/2024] Open
Abstract
Obesity is a growing concern. 42.3% of people in the U.S were considered obese between 2017-2018. Much is still unknown about the genetic components that contribute to weight gain. In humans, the hormone glucagon is a major contributor to the body's energy regulation as it signals for the breakdown of lipids. Treatments targeting the glucagon pathway have helped patients with both weight loss and appetite suppression. Understanding the genetic modifiers of glucagon signaling and its downstream pathways could enable the development of a wider variety of effective therapeutics. In this study, we blocked the glucagon pathway in Drosophila melanogaster by reducing the expression of the fly ortholog of the glucagon receptor (AKHR). We then crossed our model to the Drosophila Genetic Reference Panel (DGRP) and looked for natural variation in fat content. We used variation in larval density to identify candidate modifier genes through a genome-wide association study. We then tested these modifier genes by increasing or decreasing their expression in the AKHR model. We screened these candidates initially with the same density assay used in the original study to narrow down to four candidate genes that substantially impacted the density of the larvae: THADA, AmyD, GluRIIC, and CG9826. We further characterized these candidates using biochemical assays to analyze stored metabolites such as triglycerides, glucose, glycogen, and protein under control, high sugar, and high fat conditions to see if the larvae are resistant to environmental changes. Our results indicate consistency between the results of the density assay and direct measurement of metabolite levels. In particular, THADA and AmyD are highlighted as interesting genes for additional study. We hope to improve our understanding of the glucagon signaling pathway, obesity, and lipid metabolism. We also aim to provide candidate genes that can be regarded as future therapeutic targets.
Collapse
Affiliation(s)
- Audrey Nicol
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Malaika Ahmed
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Chelsea Fischer
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - John G. Garces
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Shana Magnus
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Nay Maung
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Nicholas Molisani
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Sophia Petrov
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Rebecca A. S. Palu
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| |
Collapse
|
15
|
Halim SA, Lodhi HW, Waqas M, Khalid A, Abdalla AN, Khan A, Al-Harrasi A. Targeting α-amylase enzyme through multi-fold structure-based virtual screening and molecular dynamic simulation. J Biomol Struct Dyn 2024; 42:5617-5630. [PMID: 37378513 DOI: 10.1080/07391102.2023.2227721] [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: 05/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023]
Abstract
α-Amylase play important role in hydrolyses of α-bonds of large α-linked polysaccharides; thus, it is a potential drug target in diabetes mellites (DM) and its inhibition is one of the therapeutic strategies in DM. With the aim to discover novel and safer therapeutic molecules to combat diabetes, a huge dataset of ∼0.69 billion compounds from ZINC20 database were screened against α-amylase using multi-fold structure-based virtual screening protocol. Based on receptor-based pharmacophore model, docking results, pharmacokinetic profile, molecular interactions with α-amylase, several compounds were retrieved as lead candidates to be further scrutinized in the in vitro assay and in vivo animal testing. Among the selected hits, CP26 exhibited the highest binding free energy in MMGB-SA analysis, followed by CP7 and CP9, which is higher than the binding free energy of acarbose. While CP20 and CP21 showed comparative binding free energy to acarbose. All the selected ligands showed acceptable binding energy range, therefore, several molecules with enhanced efficacy can be designed by derivatizing these molecules. The in-silico results indicates that the selected molecules could serve as potential selective α-amylase inhibitors and can be used for the treatment of diabetes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | | | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
16
|
Visvanathan R, Houghton MJ, Barber E, Williamson G. Structure-function relationships in (poly)phenol-enzyme binding: Direct inhibition of human salivary and pancreatic α-amylases. Food Res Int 2024; 188:114504. [PMID: 38823880 DOI: 10.1016/j.foodres.2024.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
(Poly)phenols inhibit α-amylase by directly binding to the enzyme and/or by forming starch-polyphenol complexes. Conventional methods using starch as the substrate measure inhibition from both mechanisms, whereas the use of shorter oligosaccharides as substrates exclusively measures the direct interaction of (poly)phenols with the enzyme. In this study, using a chromatography-based method and a short oligosaccharide as the substrate, we investigated the detailed structural prerequisites for the direct inhibition of human salivary and pancreatic α-amylases by over 50 (poly)phenols from the (poly)phenol groups: flavonols, flavones, flavanones, flavan-3-ols, polymethoxyflavones, isoflavones, anthocyanidins and phenolic acids. Despite being structurally very similar (97% sequence homology), human salivary and pancreatic α-amylases were inhibited to different extents by the tested (poly)phenols. The most potent human salivary α-amylase inhibitors were luteolin and pelargonidin, while the methoxylated anthocyanidins, peonidin and petunidin, significantly blocked pancreatic enzyme activity. B-ring methoxylation of anthocyanidins increased inhibition against both human α-amylases while hydroxyl groups at C3 and B3' acted antagonistically in human salivary inhibition. C4 carbonyl reduction, or the positive charge on the flavonoid structure, was the key structural feature for human pancreatic inhibition. B-ring glycosylation did not affect salivary enzyme inhibition, but increased pancreatic enzyme inhibition when compared to its corresponding aglycone. Overall, our findings indicate that the efficacy of interaction with human α-amylase is mainly influenced by the type and placement of functional groups rather than the number of hydroxyl groups and molecular weight.
Collapse
Affiliation(s)
- Rizliya Visvanathan
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia; Victorian Heart Institute, Monash University, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Michael J Houghton
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia; Victorian Heart Institute, Monash University, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Elizabeth Barber
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia; Victorian Heart Institute, Monash University, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Gary Williamson
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia; Victorian Heart Institute, Monash University, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia.
| |
Collapse
|
17
|
Chahal S, Rani P, Shweta, Goel KK, Joshi G, Singh R, Kumar P, Singh D, Sindhu J. Pyrano[2,3-c]pyrazole fused spirooxindole-linked 1,2,3-triazoles as antioxidant agents: Exploring their utility in the development of antidiabetic drugs via inhibition of α-amylase and DPP4 activity. Bioorg Chem 2024; 147:107363. [PMID: 38657527 DOI: 10.1016/j.bioorg.2024.107363] [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: 01/17/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
Environment-benign, multicomponent synthetic methodologies are vital in modern pharmaceutical research and facilitates multi-targeted drug development via synergistic approach. Herein, we reported green and efficient synthesis of pyrano[2,3-c]pyrazole fused spirooxindole linked 1,2,3-triazoles using a tea waste supported copper catalyst (TWCu). The synthetic approach involves a one-pot, five-component reaction using N-propargylated isatin, hydrazine hydrate, ethyl acetoacetate, malononitrile/ethyl cyanoacetate and aryl azides as model substrates. Mechanistically, the reaction was found to proceed via in situ pyrazolone formation followed by Knoevenagel condensation, azide alkyne cycloaddition and Michael's addition reactions. The molecules were developed using structure-based drug design. The primary goal is to identifying anti-oxidant molecules with potential ability to modulate α-amylase and DPP4 (dipeptidyl-peptidase 4) activity. The anti-oxidant analysis, as determined via DPPH, suggested that the synthesized compounds, A6 and A10 possessed excellent anti-oxidant potential compared to butylated hydroxytoluene (BHT). In contrast, compounds A3, A5, A8, A9, A13, A15, and A18 were found to possess comparable anti-oxidant potential. Among these, A3 and A13 possessed potential α-amylase inhibitory activity compared to the acarbose, and A3 further emerged as dual inhibitors of both DPP4 and α-amylase with anti-oxidant potential. The relationship of functionalities on their anti-oxidant and enzymatic inhibition was explored in context to their SAR that was further corroborated using in silico techniques and enzyme kinetics.
Collapse
Affiliation(s)
- Sandhya Chahal
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Payal Rani
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Shweta
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to be University), Haridwar 249404, India
| | - Gaurav Joshi
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal (A Central) University, Srinagar-246174, Dist. Garhwal, Uttarakhand, India
| | - Rajvir Singh
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119, India.
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak 124001, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India.
| |
Collapse
|
18
|
Mai TT, Phan MH, Thai TT, Lam TP, Lai NVT, Nguyen TT, Nguyen TVP, Vo CVT, Thai KM, Tran TD. Discovery of novel flavonoid derivatives as potential dual inhibitors against α-glucosidase and α-amylase: virtual screening, synthesis, and biological evaluation. Mol Divers 2024; 28:1629-1650. [PMID: 37369956 DOI: 10.1007/s11030-023-10680-0] [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: 01/28/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
Diabetes mellitus is one of the top ten causes of death worldwide, accounting for 6.7 million deaths in 2021, and is one of the most rapidly growing global health emergencies of this century. Although several classes of therapeutic drugs have been invented and applied in clinical practice, diabetes continues to pose a serious and growing threat to public health and places a tremendous burden on those affected and their families. The strategy of reducing carbohydrate digestibility by inhibiting the activities of α-glucosidase and α-amylase is regarded as a promising preventative treatment for type 2 diabetes. In this study, we investigated the dual inhibitory effect against two polysaccharide hydrolytic enzymes of flavonoid derivatives from an in-house chemical database. By combining molecular docking and structure-activity relationship analysis, twelve compounds with docking energies less than or equal to - 8.0 kcal mol-1 and containing required structural features for dual inhibition of the two enzymes were identified and subjected to chemical synthesis and in vitro evaluation. The obtained results showed that five compounds exhibited dual inhibitory effects on the target enzymes with better IC50 values than the approved positive control acarbose. Molecular dynamics simulations were performed to elucidate the binding of these flavonoids to the enzymes. The predicted pharmacokinetic and toxicological properties suggest that these compounds are viable for further development as type 2 diabetes drugs.
Collapse
Affiliation(s)
- Tan Thanh Mai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Minh-Hoang Phan
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thao Thi Thai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thua-Phong Lam
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Nghia Vo-Trong Lai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Thao Nguyen
- Faculty of Medicine and Pharmacy, Tay Nguyen University, Buon Ma Thuot, Dak Lak, 630000, Vietnam
| | - Thuy-Viet-Phuong Nguyen
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Cam-Van Thi Vo
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Khac-Minh Thai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Dao Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
19
|
Senger MR, da Costa Latgé SG, von Ranke NL, de Aquino GAS, Dantas RF, Genta FA, Ferreira SB, Junior FPS. Kinetics and molecular modeling studies on the inhibition mechanism of GH13 α-glycosidases by small molecule ligands. Int J Biol Macromol 2024; 269:132036. [PMID: 38697429 DOI: 10.1016/j.ijbiomac.2024.132036] [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: 02/23/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
Alpha-glucosidase inhibitors play an important role in Diabetes Mellitus (DM) treatment since they prevent postprandial hyperglycemia. The Glycoside Hydrolase family 13 (GH13) is the major family of enzymes acting on substrates containing α-glucoside linkages, such as maltose and amylose/amylopectin chains in starch. Previously, our group identified glycoconjugate 1H-1,2,3-triazoles (GCTs) inhibiting two GH13 α-glycosidases: yeast maltase (MAL12) and porcine pancreatic amylase (PPA). Here, we combined kinetic studies and computational methods on nine GCTs to characterize their inhibitory mechanism. They all behaved as reversible inhibitors, and kinetic models encompassed noncompetitive and various mechanisms of mixed-type inhibition for both enzymes. Most potent inhibitors displayed Ki values of 30 μM for MAL12 (GPESB16) and 37 μM for PPA (GPESB15). Molecular dynamics and docking simulations indicated that on MAL12, GPESB15 and GPESB16 bind in a cavity adjacent to the active site, while on the PPA, GPESB15 was predicted to bind at the entrance of the catalytic site. Notably, despite its putative location within the active site, the binding of GPESB15 does not obstruct the substrate's access to the cleavage site. Our study contributes to paving the way for developing novel therapeutic strategies for managing DM-2 through GH13 α-glycosidases inhibition.
Collapse
Affiliation(s)
- Mario Roberto Senger
- Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Samara Graciane da Costa Latgé
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Natalia Lidmar von Ranke
- Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Gabriel Alves Souto de Aquino
- Laboratório de Síntese Orgânica e Prospecção Biológica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Ferreira Dantas
- Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Fernando Ariel Genta
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Sabrina Baptista Ferreira
- Laboratório de Síntese Orgânica e Prospecção Biológica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Floriano Paes Silva Junior
- Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Ouahabi S, Daoudi NE, Loukili EH, Asmae H, Merzouki M, Bnouham M, Challioui A, Hammouti B, Fauconnier ML, Rhazi L, Ayerdi Gotor A, Depeint F, Ramdani M. Investigation into the Phytochemical Composition, Antioxidant Properties, and In-Vitro Anti-Diabetic Efficacy of Ulva lactuca Extracts. Mar Drugs 2024; 22:240. [PMID: 38921551 PMCID: PMC11204821 DOI: 10.3390/md22060240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
In this research, the chemical compositions of various extracts obtained from Ulva lactuca, a type of green seaweed collected from the Nador lagoon in the northern region of Morocco, were compared. Their antioxidant and anti-diabetic properties were also studied. Using GC-MS technology, the fatty acid content of the samples was analyzed, revealing that palmitic acid, eicosenoic acid, and linoleic acid were the most abundant unsaturated fatty acids present in all samples. The HPLC analysis indicated that sinapic acid, naringin, rutin, quercetin, cinnamic acid, salicylic acid, apigenin, flavone, and flavanone were the most prevalent phenolic compounds. The aqueous extract obtained by maceration showed high levels of polyphenols and flavonoids, with values of 379.67 ± 0.09 mg GAE/g and 212.11 ± 0.11 mg QE/g, respectively. This extract also exhibited an impressive ability to scavenge DPPH radicals, as indicated by its IC50 value of 0.095 ± 0.12 mg/mL. Additionally, the methanolic extract obtained using the Soxhlet method demonstrated antioxidant properties by preventing β-carotene discoloration, with an IC50 of 0.087 ± 0.14 mg/mL. Results from in-vitro studies showed that extracts from U. lactuca were able to significantly inhibit the enzymatic activity of α-amylase and α-glucosidase. Among the various extracts, methanolic extract (S) has been identified as the most potent inhibitor, exhibiting a statistically similar effect to that of acarbose. Furthermore, molecular docking models were used to evaluate the interaction between the primary phytochemicals found in these extracts and the human pancreatic α-amylase and α-glucosidase enzymes. These findings suggest that U. lactuca extracts contain bioactive substances that are capable of reducing enzyme activity more effectively than the commercially available drug, acarbose.
Collapse
Affiliation(s)
- Safae Ouahabi
- Laboratory of Applied and Environmental Chemistry (LCAE), Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (S.O.); (H.A.); (M.M.); (A.C.); (M.R.)
| | - Nour Elhouda Daoudi
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (N.E.D.); (M.B.)
- Higher Institute of Nursing Professions and Health Techniques, Oujda 60000, Morocco
| | - El Hassania Loukili
- Euromed Research Center, Euromed Polytechnic School, Euromed University of Fes (UEMF), Fes 30000, Morocco; (E.H.L.); (B.H.)
| | - Hbika Asmae
- Laboratory of Applied and Environmental Chemistry (LCAE), Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (S.O.); (H.A.); (M.M.); (A.C.); (M.R.)
| | - Mohammed Merzouki
- Laboratory of Applied and Environmental Chemistry (LCAE), Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (S.O.); (H.A.); (M.M.); (A.C.); (M.R.)
| | - Mohamed Bnouham
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (N.E.D.); (M.B.)
| | - Allal Challioui
- Laboratory of Applied and Environmental Chemistry (LCAE), Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (S.O.); (H.A.); (M.M.); (A.C.); (M.R.)
| | - Belkheir Hammouti
- Euromed Research Center, Euromed Polytechnic School, Euromed University of Fes (UEMF), Fes 30000, Morocco; (E.H.L.); (B.H.)
| | - Marie-Laure Fauconnier
- Laboratory of Chemistry of Natural Molecules, University of Liège, Gembloux Agro-Bio Tech. 2, Passage des Déportés, B-5030 Gembloux, Belgium;
| | - Larbi Rhazi
- Institut Polytechnique UniLaSalle, Université d’Artois, ULR 7519, UniLaSalle, 19 rue Pierre Waguet, BP 30313, 60026 Beauvais, France;
| | - Alicia Ayerdi Gotor
- Institut Polytechnique UniLaSalle, AGHYLE, UP 2018.C101, UniLaSalle, 19 rue Pierre Waguet, BP 30313, 60026 Beauvais, France;
| | - Flore Depeint
- Institut Polytechnique UniLaSalle, Université d’Artois, ULR 7519, UniLaSalle, 19 rue Pierre Waguet, BP 30313, 60026 Beauvais, France;
| | - Mohammed Ramdani
- Laboratory of Applied and Environmental Chemistry (LCAE), Faculty of Sciences, Mohammed First University, B.P. 717, Oujda 60000, Morocco; (S.O.); (H.A.); (M.M.); (A.C.); (M.R.)
| |
Collapse
|
21
|
Farazi M, Houghton MJ, Cardoso BR, Murray M, Williamson G. Inhibitory effect of extracts from edible parts of nuts on α-amylase activity: a systematic review. Food Funct 2024; 15:5209-5223. [PMID: 38717256 DOI: 10.1039/d4fo00414k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Elevated blood glucose concentration is a risk factor for developing metabolic dysfunction and insulin resistance, leading to type 2 diabetes and cardiovascular diseases. Nuts have the potential to inhibit α-amylase activity, and so lower postprandial glucose, due to their content of polyphenols and other bioactive compounds. We conducted a systematic literature review to assess the ability of extracts from commonly consumed edible parts of nuts to inhibit α-amylase. Among the 31 included papers, only four utilised human α-amylases. These papers indicated that polyphenol-rich chestnut skin extracts exhibited strong inhibition of both human salivary and pancreatic α-amylases, and that a polyphenol-rich almond skin extract was a potent inhibitor of human salivary α-amylase. The majority of the reviewed studies utilised porcine pancreatic α-amylase, which has ∼86% sequence homology with the corresponding human enzyme but with some key amino acid variations located within the active site. Polyphenol-rich extracts from chestnut, almond, kola nut, pecan and walnut, and peptides isolated from cashew, inhibited porcine pancreatic α-amylase. Some studies used α-amylases sourced from fungi or bacteria, outcomes from which are entirely irrelevant to human health, as they have no sequence homology with the human enzyme. Given the limited research involving human α-amylases, and the differences in inhibition compared to porcine enzymes and especially enzymes from microorganisms, it is recommended that future in vitro experiments place greater emphasis on utilising enzymes sourced from humans to facilitate a reliable prediction of effects in intervention studies.
Collapse
Affiliation(s)
- Mena Farazi
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia.
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Michael J Houghton
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia.
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Barbara R Cardoso
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia.
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Margaret Murray
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia.
- School of Health Sciences, Swinburne University of Technology, John St, Hawthorn, VIC 3122, Australia
| | - Gary Williamson
- Department of Nutrition, Dietetics and Food, Monash University, BASE Facility, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia.
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, VIC 3168, Australia
| |
Collapse
|
22
|
Mehmood H, Haroon M, Akhtar T, Woodward S, Haq S, M Alshehri S. Synthesis, anti-diabetic profiling and molecular docking studies of 2-(2-arylidenehydrazinyl)thiazol-4(5 H)-ones. Future Med Chem 2024; 16:1255-1266. [PMID: 38989987 PMCID: PMC11244696 DOI: 10.1080/17568919.2024.2342700] [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: 02/18/2024] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Aim: To synthesize novel more potent anti-diabetic agents. Methodology: A simple cost effective Hantzsch's synthetic strategy was used to synthesize 2-(2-arylidenehydrazinyl)thiazol-4(5H)-ones. Results: Fifteen new 2-(2-arylidenehydrazinyl)thiazol-4(5H)-ones were established to check their anti-diabetic potential. From alpha(α)-amylase inhibition, anti-glycation and anti-oxidant activities it is revealed that most of the compounds possess good anti-diabetic potential. All tested compounds were found to be more potent anti-diabetic agents via anti-glycation mode. The results of α-amylase and anti-oxidant inhibition revealed that compounds are less active against α-amylase and anti-oxidant assays. Conclusion: This study concludes that introduction of various electron withdrawing groups at the aryl ring and substitution of different functionalities around thiazolone nucleus could help to find out better anti-diabetic drug.
Collapse
Affiliation(s)
- Hasnain Mehmood
- Department of Chemistry, Mirpur University of Science & Technology (MUST), 10250, Mirpur (AJK), Pakistan
| | - Muhammad Haroon
- Department of Chemistry & Biochemistry, Miami University 651, E. High Street, Oxford, 45056, Ohio, USA
| | - Tashfeen Akhtar
- Department of Chemistry, Mirpur University of Science & Technology (MUST), 10250, Mirpur (AJK), Pakistan
| | - Simon Woodward
- GSK, Carbon Neutral Laboratories for Sustainable Chemistry, University Park Nottingham, NG7 2RD, United Kingdom
| | - Saadia Haq
- Institute of Chemistry, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, 64200, Pakistan
- Centre for Theoretical & Computational Research, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, 64200, Pakistan
| | - Saad M Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
23
|
Ma K, Su ZY, Cheng YH, Yang XP. A new α-amylase inhibitory peptide from Gynura medica extract. Food Chem 2024; 438:137959. [PMID: 37979256 DOI: 10.1016/j.foodchem.2023.137959] [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: 04/03/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
In this study, we discovered a novel peptide, Gymepeptide A, with α-amylase inhibitory activity in the water extract of Gynura medica. The structure of Gymepeptide A was determined as CGDREETR using HR-MS, 1H NMR, 13C NMR, and 2D-NMR techniques. Notably, Gymepeptide A possesses a rare double arginine residue structure and exhibits strong α-amylase inhibitory activity. Enzyme dynamic assays, molecular docking experiments, and isothermal titration calorimetry indicated that the double arginine residue structure of Gymepeptide A interacts with amino acid residues in the nearby active site region of α-amylase through hydrogen bonds and van der Waals forces. This interaction effectively inhibits the hydrolysis activity of α-amylase. Furthermore, in vitro starch digestion tests revealed that Gymepeptide A significantly reduced the digestion rate of starch and the concentration of glucose produced after starch digestion. These findings highlight the great potential of Gymepeptide A in decreasing postprandial blood glucose levels.
Collapse
Affiliation(s)
- Ke Ma
- School of Food and Biological Engineering, Key Laboratory of Industrial Biotechnology in Tobacco Industry, Zhengzhou University of Light Industry, 136 Kexuedadao Road, 450008 Zhengzhou, China
| | - Ze-Yu Su
- School of Food and Biological Engineering, Key Laboratory of Industrial Biotechnology in Tobacco Industry, Zhengzhou University of Light Industry, 136 Kexuedadao Road, 450008 Zhengzhou, China
| | - Yuan-Hang Cheng
- School of Food and Biological Engineering, Key Laboratory of Industrial Biotechnology in Tobacco Industry, Zhengzhou University of Light Industry, 136 Kexuedadao Road, 450008 Zhengzhou, China
| | - Xue-Peng Yang
- School of Food and Biological Engineering, Key Laboratory of Industrial Biotechnology in Tobacco Industry, Zhengzhou University of Light Industry, 136 Kexuedadao Road, 450008 Zhengzhou, China.
| |
Collapse
|
24
|
Han L, Zhao D, Li Y, Jin J, El-Kott AF, Al-Saeed FA, Eldib AM. Assessment of the Anti-Breast Cancer Effects of Urolithin with Molecular Docking Studies in the In Vitro Condition: Introducing a Novel Chemotherapeutic Drug. Mol Biotechnol 2024; 66:554-566. [PMID: 37280483 DOI: 10.1007/s12033-023-00766-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023]
Abstract
A lot of research has been done on using natural items as diabetes treatment. The molecular docking study was conducted to evaluate the inhibitory activities of urolithin A against α-amylase, α-glucosidase, and aldose reductase. The molecular docking calculations indicated the probable interactions and the characteristics of these contacts at an atomic level. The results of the docking calculations showed the docking score of urolithin A against α-amylase was -5.169 kcal/mol. This value for α-glucosidase and aldose reductase was -3.657 kcal/mol and -7.635 kcal/mol, respectively. In general, the outcomes of the docking calculations revealed that urolithin A can construct several hydrogen bonds and hydrophobic contacts with the assessed enzymes and reduces their activities considerably. The properties of urolithin against common human breast cancer cell lines, i.e., SkBr3, MDA-MB-231, MCF-7, Hs578T, Evsa-T, BT-549, AU565 and 600MPE were evaluated. The IC50 of the urolithin was 400, 443, 392, 418, 397, 530, 566 and 551 against SkBr3, MDA-MB-231, MCF-7, Hs578T, Evsa-T, BT-549, AU565 and 600MPE, respectively. After doing the clinical trial studies, the recent molecule may be used as an anti-breast cancer supplement in humans. IC50 values of urolithin A on α-amylase, α-glucosidase, and aldose reductase enzymes were obtained at 16.14, 1.06 and 98.73 µM, respectively.
Collapse
Affiliation(s)
- Lu Han
- Department of General Surgery, Sijing Hospital of Songjiang District Shanghai, Shanghai, 201601, China
| | - Danbo Zhao
- Department of Oncology, Ezhou Central Hospital, Ezhou, 436000, Hubei, China
| | - Ya Li
- Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi, 710061, China
| | - Jianwei Jin
- Department of Oncology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Fatimah A Al-Saeed
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
| | - Ali M Eldib
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
- Alrayan Medical Colleges (AMC), Hejrah Street, P. O. Box 41411, Madinah, Kingdom of Saudi Arabia
| |
Collapse
|
25
|
Zhang L, Kong H, Chitrakar B, Ban X, Gu Z, Hong Y, Cheng L, Li Z, Li C. The substitution sites of hydroxyl and galloyl groups determine the inhibitory activity of human pancreatic α-amylase in twelve tea polyphenol monomers. Int J Biol Macromol 2024; 259:129189. [PMID: 38181909 DOI: 10.1016/j.ijbiomac.2023.129189] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
Tea polyphenols have been reported as potential α-amylase inhibitors. However, the quantitative structure-activity relationship (QSAR) between tea polyphenols and human pancreas α-amylase (HPA) is not well understood. Herein, the inhibitory effect of twelve tea polyphenol monomers on HPA was investigated in terms of inhibitory activity, as well as QSAR analysis and interaction mechanism. The results revealed that the HPA inhibitory activity of theaflavins (TFs), especially theaflavin-3'-gallate (TF-3'-G, IC50: 0.313 mg/mL), was much stronger than that of catechins (IC50: 18.387-458.932 mg/mL). The QSAR analysis demonstrated that the determinant for the inhibitory activity of HPA was not the number of hydroxyl and galloyl groups in tea polyphenol monomers, while the substitution sites of these groups potentially might play a more important role in modulating the inhibitory activity. The inhibition kinetics and molecular docking revealed that TF-3'-G as a mixed-type inhibitor had the lowest inhibition constant and bound to the active sites of HPA with the lowest binding energy (-7.74 kcal/mol). These findings could provide valuable insights into the structures-activity relationships between tea polyphenols and the HPA inhibitors.
Collapse
Affiliation(s)
- Lan Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haocun Kong
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bimal Chitrakar
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, Hebei, China
| | - Xiaofeng Ban
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Li Cheng
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China.
| | - Caiming Li
- Key Laboratory of Synergetic and Biological Colloids, Ministry of Education, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China.
| |
Collapse
|
26
|
Nada AA, Metwally AM, Asaad AM, Celik I, Ibrahim RS, Eldin SMS. Synergistic effect of potential alpha-amylase inhibitors from Egyptian propolis with acarbose using in silico and in vitro combination analysis. BMC Complement Med Ther 2024; 24:65. [PMID: 38291462 PMCID: PMC10826043 DOI: 10.1186/s12906-024-04348-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Type 2 Diabetes mellitus (DM) is an affliction impacting the quality of life of millions of people worldwide. An approach used in the management of Type 2 DM involves the use of the carbohydrate-hydrolyzing enzyme inhibitor, acarbose. Although acarbose has long been the go-to drug in this key approach, it has become apparent that its side effects negatively impact patient adherence and subsequently, therapeutic outcomes. Similar to acarbose in its mechanism of action, bee propolis, a unique natural adhesive biomass consisting of biologically active metabolites, has been found to have antidiabetic potential through its inhibition of α-amylase. To minimize the need for ultimately novel agents while simultaneously aiming to decrease the side effects of acarbose and enhance its efficacy, combination drug therapy has become a promising pharmacotherapeutic strategy and a focal point of this study. METHODS Computer-aided molecular docking and molecular dynamics (MD) simulations accompanied by in vitro testing were used to mine novel, pharmacologically active chemical entities from Egyptian propolis to combat Type 2 DM. Glide docking was utilized for a structure-based virtual screening of the largest in-house library of Egyptian propolis metabolites gathered from literature, in addition to GC-MS analysis of the propolis sample under investigation. Thereafter, combination analysis by means of fixed-ratio combinations of acarbose with propolis and the top chosen propolis-derived phytoligand was implemented. RESULTS Aucubin, identified for the first time in propolis worldwide and kaempferol were the most promising virtual hits. Subsequent in vitro α-amylase inhibitory assay demonstrated the ability of these hits to significantly inhibit the enzyme in a dose-dependent manner with an IC50 of 2.37 ± 0.02 mM and 4.84 ± 0.14 mM, respectively. The binary combination of acarbose with each of propolis and kaempferol displayed maximal synergy at lower effect levels. Molecular docking and MD simulations revealed a cooperative binding mode between kaempferol and acarbose within the active site. CONCLUSION The suggested strategy seems imperative to ensure a steady supply of new therapeutic entities sourced from Egyptian propolis to regress the development of DM. Further pharmacological in vivo investigations are required to confirm the potent antidiabetic potential of the studied combination.
Collapse
Affiliation(s)
- Ahmed A Nada
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alkhartoom Square, Alexandria, 21521, Egypt
| | - Aly M Metwally
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alkhartoom Square, Alexandria, 21521, Egypt
| | - Aya M Asaad
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alkhartoom Square, Alexandria, 21521, Egypt
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, 38039, Turkey
| | - Reham S Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alkhartoom Square, Alexandria, 21521, Egypt.
| | - Safa M Shams Eldin
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alkhartoom Square, Alexandria, 21521, Egypt
| |
Collapse
|
27
|
Khan KM, Nadeem MF, Mannan A, Chohan TA, Islam M, Ansari SA, Alkahtani HM, Ansari IA, Khurshid U, Abidin SAZ, Shah SAA, Saleem H. Biochemical, Toxicological, and in Silico Aspects of Trillium govanianum Wall. ex D.Don (Trilliaceae): A Rich Source of Natural Bioactive Compounds. Chem Biodivers 2024; 21:e202301375. [PMID: 38031244 DOI: 10.1002/cbdv.202301375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/01/2023]
Abstract
Trillium govanianum is a high-value medicinal herb, having multifunctional traditional and culinary uses. The present investigation was carried out to evaluate the phytochemical, biological and toxicological parameters of the T. govanianum Wall. ex D. Don (Family: Trilliaceae) roots collected from Azad Kashmir, Pakistan. Phytochemical profiling was achieved by determining total bioactive contents (total phenolic and flavonoid contents) and UHPLC-MS analysis. For biological evaluation, antioxidant activities (DPPH, ABTS, FRAP, CUPRAC, phosphomolybdenum, and metal chelation assays) and enzyme inhibition activities (against AChE, BChE, glucosidase, amylase, and tyrosinase) were performed. Moreover, cytotoxicity was assessed against three human carcinoma cell lines (MDA-MB-231, CaSki, and DU-145). The tested extract was found to contain higher total phenolics (7.56 mg GAE/g dry extract) as compared to flavonoid contents (0.45 mg RE/g dry extract). Likewise, for the antioxidant activity, higher CUPRAC activity was noted with 39.84 mg TE/g dry extract values. In the case of enzyme assays, higher activity was pointed out against the cholinesterase, glucosidase and tyrosinase enzymes. The plant extract displayed significant cytotoxicity against the cell lines examined. Moreover, the in-silico studies highlighted the interaction between the important phytochemicals and tested enzymes. To conclude, the assessed biological activity and the existence of bioactive phytochemicals in the studied plant extract may pave the way for the development of novel pharmaceuticals.
Collapse
Affiliation(s)
- Kashif Maqbool Khan
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Muhammad Faisal Nadeem
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Abdul Mannan
- Department of Pharmacy, COMSATS Institute of Information Technology, Abbottabad, 22060, Pakistan
| | - Tahir Ali Chohan
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Muhammad Islam
- University College of Pharmacy, Punjab University, Pakistan
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P, .O Box 2457, Riyadh, 11451, Saudi Arabia
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P, .O Box 2457, Riyadh, 11451, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, 10124, Turin, Italy
| | - Umair Khurshid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, 47500, Bandar Sunway Selangor Darul Ehsan, Malaysia
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor, 42300, Malaysia
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Hammad Saleem
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| |
Collapse
|
28
|
Ashok PP, Dasgupta D, Ray A, Suman SK. Challenges and prospects of microbial α-amylases for industrial application: a review. World J Microbiol Biotechnol 2023; 40:44. [PMID: 38114825 DOI: 10.1007/s11274-023-03821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 12/21/2023]
Abstract
α-Amylases are essential biocatalysts representing a billion-dollar market with significant long-term global demand. They have varied applications ranging from detergent, textile, and food sectors such as bakery to, more recently, biofuel industries. Microbial α-amylases have distinct advantages over their plant and animal counterparts owing to generally good activities and better stability at temperature and pH extremes. With the scope of applications expanding, the need for new and improved α-amylases is ever-growing. However, scaling up microbial α-amylase technology from the laboratory to industry for practical applications is impeded by several issues, ranging from mass transfer limitations, low enzyme yields, and energy-intensive product recovery that adds to high production costs. This review highlights the major challenges and prospects for the production of microbial α-amylases, considering the various avenues of industrial bioprocessing such as culture-independent approaches, nutrient optimization, bioreactor operations with design improvements, and product down-streaming approaches towards developing efficient α-amylases with high activity and recyclability. Since the sequence and structure of the enzyme play a crucial role in modulating its functional properties, we have also tried to analyze the structural composition of microbial α-amylase as a guide to its thermodynamic properties to identify the areas that can be targeted for enhancing the catalytic activity and thermostability of the enzyme through varied immobilization or selective enzyme engineering approaches. Also, the utilization of inexpensive and renewable substrates for enzyme production to isolate α-amylases with non-conventional applications has been briefly discussed.
Collapse
Affiliation(s)
- Patel Pratima Ashok
- Biochemistry and Biotechnology Area, Material Resource Efficiency Division, CSIR-Indian Institute of Petroleum, Dehradun, 248005, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Diptarka Dasgupta
- Biochemistry and Biotechnology Area, Material Resource Efficiency Division, CSIR-Indian Institute of Petroleum, Dehradun, 248005, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Anjan Ray
- Biochemistry and Biotechnology Area, Material Resource Efficiency Division, CSIR-Indian Institute of Petroleum, Dehradun, 248005, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sunil K Suman
- Biochemistry and Biotechnology Area, Material Resource Efficiency Division, CSIR-Indian Institute of Petroleum, Dehradun, 248005, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
29
|
Raut B, Upadhyaya SR, Bashyal J, Parajuli N. In Silico and In Vitro Analyses to Repurpose Quercetin as a Human Pancreatic α-Amylase Inhibitor. ACS OMEGA 2023; 8:43617-43631. [PMID: 38027372 PMCID: PMC10666247 DOI: 10.1021/acsomega.3c05082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/20/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Human pancreatic α-amylase (HPA), situated at the apex of the starch digestion hierarchy, is an attractive therapeutic approach to precisely regulate blood glucose levels, thereby efficiently managing diabetes. Polyphenols offer a natural and multifaceted approach to moderate postprandial sugar spikes, with their slight modulation in carbohydrate digestion and potential secondary benefits, such as antioxidant and anti-inflammatory effects. Taking into consideration the unfavorable side effects of currently available commercial medications, we aimed to study a library of polyphenols attributed to their remarkable antidiabetic properties and screened the most potent HPA inhibitor via a comprehensive in silico study encompassing molecular docking, molecular mechanics with generalized Born and surface area solvation (MM/GBSA) calculation, molecular dynamics (MD) simulation, density functional theory (DFT) study, and pharmacokinetic properties followed by an in vitro assay. Significant hydrogen bonding with the catalytic triad residues of HPA, prominent MM/GBSA binding energy of -27.03 kcal/mol, and the stable nature of the protein-ligand complex with regard to 100 ns MD simulation screened quercetin as the best HPA inhibitor. Additionally, quercetin showed strong reactivity in the substrate-binding pocket of HPA and exhibited favorable pharmacokinetic properties with a considerable inhibitory concentration (IC50) of 57.37 ± 0.9 μg/mL against α-amylase. This study holds prospects for HPA inhibition and suggests quercetin as an approach to therapy for diabetes; however, it is imperative to conduct further research.
Collapse
Affiliation(s)
- Bimal
K. Raut
- Central Department of Chemistry, Tribhuvan University, Kirtipur 44600, Kathmandu, Nepal
| | - Siddha Raj Upadhyaya
- Central Department of Chemistry, Tribhuvan University, Kirtipur 44600, Kathmandu, Nepal
| | - Jyoti Bashyal
- Central Department of Chemistry, Tribhuvan University, Kirtipur 44600, Kathmandu, Nepal
| | - Niranjan Parajuli
- Central Department of Chemistry, Tribhuvan University, Kirtipur 44600, Kathmandu, Nepal
| |
Collapse
|
30
|
Kalinovskii AP, Sintsova OV, Gladkikh IN, Leychenko EV. Natural Inhibitors of Mammalian α-Amylases as Promising Drugs for the Treatment of Metabolic Diseases. Int J Mol Sci 2023; 24:16514. [PMID: 38003703 PMCID: PMC10671682 DOI: 10.3390/ijms242216514] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
α-Amylase is a generally acknowledged molecular target of a distinct class of antidiabetic drugs named α-glucosidase inhibitors. This class of medications is scarce and rather underutilized, and treatment with current commercial drugs is accompanied by unpleasant adverse effects. However, mammalian α-amylase inhibitors are abundant in nature and form an extensive pool of high-affinity ligands that are available for drug discovery. Individual compounds and natural extracts and preparations are promising therapeutic agents for conditions associated with impaired starch metabolism, e.g., diabetes mellitus, obesity, and other metabolic disorders. This review focuses on the structural diversity and action mechanisms of active natural products with inhibitory activity toward mammalian α-amylases, and emphasizes proteinaceous inhibitors as more effective compounds with significant potential for clinical use.
Collapse
Affiliation(s)
- Aleksandr P. Kalinovskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Oksana V. Sintsova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia; (O.V.S.); (I.N.G.)
| | - Irina N. Gladkikh
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia; (O.V.S.); (I.N.G.)
| | - Elena V. Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia; (O.V.S.); (I.N.G.)
| |
Collapse
|
31
|
Kim Y, Li H, Choi J, Boo J, Jo H, Hyun JY, Shin I. Glycosidase-targeting small molecules for biological and therapeutic applications. Chem Soc Rev 2023; 52:7036-7070. [PMID: 37671645 DOI: 10.1039/d3cs00032j] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Glycosidases are ubiquitous enzymes that catalyze the hydrolysis of glycosidic linkages in oligosaccharides and glycoconjugates. These enzymes play a vital role in a wide variety of biological events, such as digestion of nutritional carbohydrates, lysosomal catabolism of glycoconjugates, and posttranslational modifications of glycoproteins. Abnormal glycosidase activities are associated with a variety of diseases, particularly cancer and lysosomal storage disorders. Owing to the physiological and pathological significance of glycosidases, the development of small molecules that target these enzymes is an active area in glycoscience and medicinal chemistry. Research efforts carried out thus far have led to the discovery of numerous glycosidase-targeting small molecules that have been utilized to elucidate biological processes as well as to develop effective chemotherapeutic agents. In this review, we describe the results of research studies reported since 2018, giving particular emphasis to the use of fluorescent probes for detection and imaging of glycosidases, activity-based probes for covalent labelling of these enzymes, glycosidase inhibitors, and glycosidase-activatable prodrugs.
Collapse
Affiliation(s)
- Yujun Kim
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Hui Li
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Joohee Choi
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Jihyeon Boo
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Hyemi Jo
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| |
Collapse
|
32
|
Calvert M, Sweeney RP, Chen HM, Bajwa H, Nasseri SA, Habibi D, Withers SG. Branched montbretin A mimics allow derivatisation and potent amylase inhibition. Org Biomol Chem 2023; 21:7977-7983. [PMID: 37751159 DOI: 10.1039/d3ob01051a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Mimics of the complex flavonol glycoside montbretin A in which a flavonol moiety is coupled to a caffeic acid via partially peptidic linkers have proved to be potent inhibitors of human pancreatic alpha-amylase with potential as therapeutics for control of blood glucose levels. After exploring optimal linker length, a synthetic route to a version with a branched linker was devised based on the structure of the enzyme/inhibitor complex. The resultant branched inhibitors were shown to retain nanomolar potency even when decorated with polymers as a means of modifying solubility. Similar improvements, along with nanomolar affinity, could also be achieved through conjugation to cyclodextrins which have the potential to bind to starch binding sites found on the surface of human amylase. Incorporation of a conjugatable branch into this unusual pharmacophore thereby affords considerable flexibility for further modifications to improve pharmacokinetic behaviour or as a site for attachment of capture tags or fluorophores.
Collapse
Affiliation(s)
- Matthew Calvert
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C., V6T 1Z1, Canada.
| | - Ryan P Sweeney
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C., V6T 1Z1, Canada.
| | - Hong-Ming Chen
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C., V6T 1Z1, Canada.
| | - Harbir Bajwa
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C., V6T 1Z1, Canada.
| | - Seyed A Nasseri
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C., V6T 1Z1, Canada.
| | - Davood Habibi
- Department of Organic Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, 6517838683, Iran
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C., V6T 1Z1, Canada.
| |
Collapse
|
33
|
Kashtoh H, Baek KH. New Insights into the Latest Advancement in α-Amylase Inhibitors of Plant Origin with Anti-Diabetic Effects. PLANTS (BASEL, SWITZERLAND) 2023; 12:2944. [PMID: 37631156 PMCID: PMC10458243 DOI: 10.3390/plants12162944] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023]
Abstract
The rising predominance of type 2 diabetes, combined with the poor medical effects seen with commercially available anti-diabetic medications, has motivated the development of innovative treatment approaches for regulating postprandial glucose levels. Natural carbohydrate digestion enzyme inhibitors might be a viable option for blocking dietary carbohydrate absorption with fewer side effects than manufactured medicines. Alpha-amylase is a metalloenzyme that facilitates digestion by breaking down polysaccharides into smaller molecules such as maltose and maltotriose. It also contributes to elevated blood glucose levels and postprandial hyperglycemia. As a result, scientists are being urged to target α-amylase and create inhibitors that can slow down the release of glucose from carbohydrate chains and prolong its absorption, thereby resulting in lower postprandial plasma glucose levels. Natural α-amylase inhibitors derived from plants have gained popularity as safe and cost-effective alternatives. The bioactive components responsible for the inhibitory actions of various plant extracts have been identified through phytochemical research, paving the way for further development and application. The majority of the findings, however, are based on in vitro investigations. Only a few animal experiments and very few human investigations have confirmed these findings. Despite some promising results, additional investigation is needed to develop feasible anti-diabetic drugs based on plant-derived pancreatic α-amylase inhibitors. This review summarizes the most recent findings from research on plant-derived pancreatic α-amylase inhibitors, including plant extracts and plant-derived bioactive compounds. Furthermore, it offers insights into the structural aspects of the crucial therapeutic target, α-amylases, in addition to their interactions with inhibitors.
Collapse
Affiliation(s)
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
34
|
Rhimi M, Da Lage JL, Haser R, Feller G, Aghajari N. Structural and Functional Characterization of Drosophila melanogaster α-Amylase. Molecules 2023; 28:5327. [PMID: 37513201 PMCID: PMC10384113 DOI: 10.3390/molecules28145327] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Insects rely on carbohydrates such as starch and glycogen as an energy supply for growth of larvae and for longevity. In this sense α-amylases have essential roles under extreme conditions, e.g., during nutritional or temperature stress, thereby contributing to survival of the insect. This makes them interesting targets for combating insect pests. Drosophila melanogaster α-amylase, DMA, which belongs to the glycoside hydrolase family 13, sub family 15, has been studied from an evolutionary, biochemical, and structural point of view. Our studies revealed that the DMA enzyme is active over a broad temperature and pH range, which is in agreement with the fluctuating environmental changes with which the insect is confronted. Crystal structures disclosed a new nearly fully solvated metal ion, only coordinated to the protein via Gln263. This residue is only conserved in the subgroup of D. melanogaster and may thus contribute to the enzyme adaptive response to large temperature variations. Studies of the effect of plant inhibitors and the pseudo-tetrasaccharide inhibitor acarbose on DMA activity, allowed us to underline the important role of the so-called flexible loop on activity/inhibition, but also to suggest that the inhibition modes of the wheat inhibitors WI-1 and WI-3 on DMA, are likely different.
Collapse
Affiliation(s)
- Moez Rhimi
- Molecular Microbiology and Structural Biochemistry, UMR5086, CNRS, University of Lyon 1, 7 Passage du Vercors, F-69367 Lyon, CEDEX 07, France
| | - Jean-Luc Da Lage
- Evolution, Génomes, Comportement, Ecologie, UMR 9191 University Paris-Saclay-CNRS-IRD, F-91190 Gif-sur-Yvette, France
| | - Richard Haser
- Molecular Microbiology and Structural Biochemistry, UMR5086, CNRS, University of Lyon 1, 7 Passage du Vercors, F-69367 Lyon, CEDEX 07, France
| | - Georges Feller
- Laboratory of Biochemistry, Center for Protein Engineering-InBioS, Institute of Chemistry B6a, University of Liège, B-4000 Liège, Belgium
| | - Nushin Aghajari
- Molecular Microbiology and Structural Biochemistry, UMR5086, CNRS, University of Lyon 1, 7 Passage du Vercors, F-69367 Lyon, CEDEX 07, France
| |
Collapse
|
35
|
Liu X, Sun B, Xu C, Zhang T, Zhang Y, Zhu L. Intrinsic mechanisms for the inhibition effect of graphene oxide on the catalysis activity of alpha amylase. JOURNAL OF HAZARDOUS MATERIALS 2023; 453:131389. [PMID: 37043854 DOI: 10.1016/j.jhazmat.2023.131389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/25/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
Comprehending the interactions between graphene oxide (GO) and enzymes is critical for understanding the toxicities of GO. In this study, the inherent interactions of GO with α-amylase as a typical enzyme, and the impacts of GO on the conformation and biological activities of α-amylase were systematically investigated. The results reveal that GO formed ground-state complex with α-amylase primarily via hydrogen bonding and van der Waals interactions, thus quenching the intrinsic fluorescence of the protein statically. Particularly, the strong interactions altered the microenvironment of tyrosine and tryptophan residues, caused rearrangement of polypeptide structure, and reduced the contents of α-helices and β-sheets, thus changing the conformational structure of α-amylase. According to molecular docking results, GO binds with the amino acid residues (i.e., His299, Asp300, and His305) of α-amylase mainly through hydrogen bonding, which is in accordance with in vitro incubation experiments. As a consequence, the ability of α-amylase to catalyze starch hydrolysis into glucose was depressed by GO, suggesting that GO might cause dysfunction of α-amylase. This study discloses the intrinsic binding mechanisms of GO with α-amylase and provides novel insights into the adverse effects of GO as it enters organisms.
Collapse
Affiliation(s)
- Xinwei Liu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, PR China
| | - Binbin Sun
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, PR China
| | - Chunyi Xu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, PR China
| | - Tianxu Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, PR China
| | - Yinqing Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, PR China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, PR China.
| |
Collapse
|
36
|
Kumari S, Saini R, Bhatnagar A, Mishra A. HR-LCMS and evaluation of anti-diabetic activity of Hemidesmus indicus (anantmool): Kinetic study, and molecular modelling approach. Comput Biol Chem 2023; 105:107896. [PMID: 37263051 DOI: 10.1016/j.compbiolchem.2023.107896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/08/2023] [Accepted: 05/20/2023] [Indexed: 06/03/2023]
Abstract
This study delved into the exploration of novel antidiabetic medications acquired from natural resources, utilizing the Ayurvedic Rasayana herb Hemidesmus indicus through cutting-edge chemoprofiling and molecular modelling techniques. The methanolic extract of Hemidesmus indicus root exhibited the highest extractive yield (24.70 ± 0.08 %) and contained substantial levels of total phenolic and flavonoid content as 154.15 ± 1.24 mg Gallic Acid Equivalent/g extract and 70.61 ± 0.35 Quercetin Equivalent/g extract respectively. Invitro study revealed the potent inhibitory potential of methanolic extract of the herb against essential carbohydrate hydrolytic enzymes α-amylase (IC50 = 4.19 ± 0.04 mg/ml) and α-glucosidase (IC50 = 5.78 ± 0.10 mg/ml). Further, the enzyme kinetic study demonstrated the competitive mode of inhibition of both enzymes. HR-LCMS analysis identified the major phytoconstituents present in the extracts, including Solanocapsine, Cyclovirobuxine C, Lucidine B, Zygadenine, Aspidospermidine, silychristin, 3beta-3-Hydroxy-18-lupen-21-one, Manglupenone, and 19-Noretiocholanolone. Molecular docking, molecular dynamic simulation, and MM/GBSA analysis have proved stable, rigid, compact, and folded form of complexes during the entire 100 ns simulation, illustrating Zygadenine, Solanocapsine, and Cyclovirobuxine C as the superior inhibitors of α-A protein, while Zygadenine, Plumieride, and Phlegmarine exhibited greater inhibitory behaviour towards α-G protein than the FDA-approved drug acarbose. Collectively, our findings indicate that the Hemidesmus indicus could be a promising source of α-A and α-G inhibitors, potentially serving as a lead in order to develop medications for type-2 diabetes.
Collapse
Affiliation(s)
- Sonali Kumari
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Ravi Saini
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Aditi Bhatnagar
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Abha Mishra
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
37
|
Tian Y, Wang Y, Zhong Y, Møller MS, Westh P, Svensson B, Blennow A. Interfacial Catalysis during Amylolytic Degradation of Starch Granules: Current Understanding and Kinetic Approaches. Molecules 2023; 28:molecules28093799. [PMID: 37175208 PMCID: PMC10180094 DOI: 10.3390/molecules28093799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Enzymatic hydrolysis of starch granules forms the fundamental basis of how nature degrades starch in plant cells, how starch is utilized as an energy resource in foods, and develops efficient, low-cost saccharification of starch, such as bioethanol and sweeteners. However, most investigations on starch hydrolysis have focused on its rates of degradation, either in its gelatinized or soluble state. These systems are inherently more well-defined, and kinetic parameters can be readily derived for different hydrolytic enzymes and starch molecular structures. Conversely, hydrolysis is notably slower for solid substrates, such as starch granules, and the kinetics are more complex. The main problems include that the surface of the substrate is multifaceted, its chemical and physical properties are ill-defined, and it also continuously changes as the hydrolysis proceeds. Hence, methods need to be developed for analyzing such heterogeneous catalytic systems. Most data on starch granule degradation are obtained on a long-term enzyme-action basis from which initial rates cannot be derived. In this review, we discuss these various aspects and future possibilities for developing experimental procedures to describe and understand interfacial enzyme hydrolysis of native starch granules more accurately.
Collapse
Affiliation(s)
- Yu Tian
- Department of Plant and Environmental Sciences, University of Copenhagen, DK-1871 Frederiksberg C, Denmark
| | - Yu Wang
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Yuyue Zhong
- Department of Plant and Environmental Sciences, University of Copenhagen, DK-1871 Frederiksberg C, Denmark
| | - Marie Sofie Møller
- Applied Molecular Enzyme Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Peter Westh
- Interfacial Enzymology, Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Birte Svensson
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Andreas Blennow
- Department of Plant and Environmental Sciences, University of Copenhagen, DK-1871 Frederiksberg C, Denmark
| |
Collapse
|
38
|
Mehmood H, Akhtar T, Haroon M, Khalid M, Woodward S, Asghar MA, Baby R, Orfali R, Perveen S. Synthesis of Fluorinated Hydrazinylthiazole Derivatives: A Virtual and Experimental Approach to Diabetes Management. ACS OMEGA 2023; 8:11433-11446. [PMID: 37008089 PMCID: PMC10061536 DOI: 10.1021/acsomega.3c00265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/07/2023] [Indexed: 06/19/2023]
Abstract
A novel series of fluorophenyl-based thiazoles was synthesized following the Hanztsch method. All of the compounds were initially verified with physical parameters (color, melting point, retardation factor (R f)), which were further confirmed by several spectroscopic methods, including ultraviolet-visible (UV-visible), Fourier-transform infrared (FTIR), 1H, 13C, 19F NMR, and high-resolution mass spectrometry (HRMS). The binding interactions of all compounds were studied using a molecular docking simulation approach. Furthermore, each compound was evaluated for its alpha(α)-amylase, antiglycation, and antioxidant potentials. The biocompatibility of all compounds was checked with an in vitro hemolytic assay. All synthesized scaffolds were found biocompatible with minimal lysis of human erythrocytes as compared to the standard Triton X-100. Among the tested compounds, the analogue 3h (IC50 = 5.14 ± 0.03 μM) was found to be a highly potent candidate against α-amylase as compared to the standard (acarbose, IC50 = 5.55 ± 0.06 μM). The compounds 3d, 3f, 3i, and 3k exhibited excellent antiglycation inhibition potential with their IC50 values far less than the standard amino guanidine (IC50 = 0.403 ± 0.001 mg/mL). The antidiabetic potential was further supported by docking studies. Docking studies revealed that all synthesized compounds exhibited various interactions along enzyme active sites (pi-pi, H-bonding, van der Waals) with varied binding energies.
Collapse
Affiliation(s)
- Hasnain Mehmood
- Department
of Chemistry, Mirpur University of Science
and Technology (MUST), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
| | - Tashfeen Akhtar
- Department
of Chemistry, Mirpur University of Science
and Technology (MUST), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
| | - Muhammad Haroon
- Department
of Chemistry, Mirpur University of Science
and Technology (MUST), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
- Department
of Chemistry, Government Major Muhammad Afzal Khan (Shaheed), Boys
Degree College Afzalpur, Mirpur (Affiliated
with Mirpur University of Science and Technology (MUST)), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
| | - Muhammad Khalid
- Institute
of Chemistry, Khwaja Fareed University of
Engineering & Information Technology, Rahim Yar Khan 64200, Pakistan
- Center
for Theoretical and Computational Research, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan 64200, Pakistan
| | - Simon Woodward
- GSK,
Carbon Neutral Laboratories for Sustainable Chemistry, University Park Nottingham, Nottingham NG7 2RD, U.K.
| | - Muhammad Adnan Asghar
- Department
of Chemistry, Division of Science and Technology, University of Education, Lahore 54770, Pakistan
| | - Rabia Baby
- Department
of Education, Sukkur IBA University, Sukkur 65200, Sindh, Pakistan
| | - Raha Orfali
- Department
of Pharmacognosy, Collage of Pharmacy, King
Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Shagufta Perveen
- Department
of Chemistry, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, Maryland 21251, United States
| |
Collapse
|
39
|
Basnet S, Ghimire MP, Lamichhane TR, Adhikari R, Adhikari A. Identification of potential human pancreatic α-amylase inhibitors from natural products by molecular docking, MM/GBSA calculations, MD simulations, and ADMET analysis. PLoS One 2023; 18:e0275765. [PMID: 36928801 PMCID: PMC10019617 DOI: 10.1371/journal.pone.0275765] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
Human pancreatic α-amylase (HPA), which works as a catalyst for carbohydrate hydrolysis, is one of the viable targets to control type 2 diabetes. The inhibition of α-amylase lowers blood glucose levels and helps to alleviate hyperglycemia complications. Herein, we systematically screened the potential HPA inhibitors from a library of natural products by molecular modeling. The modeling encompasses molecular docking, MM/GBSA binding energy calculations, MD simulations, and ADMET analysis. This research identified newboulaside B, newboulaside A, quercetin-3-O-β-glucoside, and sasastilboside A as the top four potential HPA inhibitors from the library of natural products, whose Glide docking scores and MM/GBSA binding energies range from -9.191 to -11.366 kcal/mol and -19.38 to -77.95 kcal/mol, respectively. Based on the simulation, among them, newboulaside B was found as the best HPA inhibitor. Throughout the simulation, with the deviation of 3Å (acarbose = 3Å), it interacted with ASP356, ASP300, ASP197, THR163, ARG161, ASP147, ALA106, and GLN63 via hydrogen bonding. Additionally, the comprehensive ADMET analysis revealed that it has good pharmacokinetic properties having not acutely toxic, moderately bioavailable, and non-inhibitor nature toward cytochrome P450. All the results suggest that newboulaside B might be a promising candidate for drug discovery against type 2 diabetes.
Collapse
Affiliation(s)
- Santosh Basnet
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | | | - Tika Ram Lamichhane
- Central Department of Physics, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | | | - Achyut Adhikari
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
40
|
Guo S, Liu S, Meng J, Gu D, Wang Y, He D, Yang Y. Dual-target affinity analysis and separation of α-amylase and α-glucosidase inhibitors from Morus alba leaves using a magnetic bifunctional immobilized enzyme system. Biomed Chromatogr 2023; 37:e5571. [PMID: 36520456 DOI: 10.1002/bmc.5571] [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: 10/07/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/16/2022]
Abstract
Morus alba leaves are a natural product with great antidiabetic potential. However, the therapeutic efficacy of natural products is usually achieved through the interaction of active compounds with specific targets. Among them, active compounds with multi-target therapeutic functions are more effective than single-target enzymes. In this study, a bienzyme system was constructed by co-immobilizing α-amylase and α-glucosidase onto Fe3 O4 for affinity screening of dual-target active components in the complex extract from M. alba leaves. As a result, a potential active compound was selectively screened by ligand fishing, separated by high-speed countercurrent chromatography using a solvent system of ethyl acetate-n-butanol-water (3:2:5, v/v), and identified as rutin. In addition, the result of molecular docking showed that rutin could interact with the active center of α-amylase and α-glucosidase through multiple hydrogen bonds, van der Waals forces, etc. to play an inhibitory role. These results demonstrate the effectiveness of the polydopamine magnetically immobilized bienzyme system for dual-target affinity screening of active substances. This study not only reveals the chemical basis of the antidiabetic activity of M. alba leaves from a dual-target perspective, but also promotes the progress of multitarget affinity screening.
Collapse
Affiliation(s)
- Shuang Guo
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, China
| | - Shuo Liu
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, China
| | - Jing Meng
- College of Marine Science and Environment, Dalian Ocean University, Dalian, China
| | - Dongyu Gu
- College of Marine Science and Environment, Dalian Ocean University, Dalian, China
| | - Yi Wang
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
| | - Dajun He
- College of Life Science, Analysis and Testing Centre, Shihezi University, Shihezi, China
| | - Yi Yang
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
41
|
In Vitro Assessment Methods for Antidiabetic Peptides from Legumes: A Review. Foods 2023; 12:foods12030631. [PMID: 36766167 PMCID: PMC9914741 DOI: 10.3390/foods12030631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Almost 65% of the human protein supply in the world originates from plants, with legumes being one of the highest contributors, comprising between 20 and 40% of the protein supply. Bioactive peptides from various food sources including legumes have been reported to show efficacy in modulating starch digestion and glucose absorption. This paper will provide a comprehensive review on recent in vitro studies that have been performed on leguminous antidiabetic peptides, focusing on the α-amylase inhibitor, α-glucosidase inhibitor, and dipeptidyl peptidase-IV (DPP-IV) inhibitor. Variations in legume cultivars and methods affect the release of peptides. Different methods have been used, such as in sample preparation, including fermentation (t, T), germination (t), and pre-cooking; in protein extraction, alkaline extraction, isoelectric precipitation, phosphate buffer extraction, and water extraction; in protein hydrolysis enzyme types and combination, enzyme substrate ratio, pH, and time; and in enzyme inhibitory assays, positive control type and concentration, inhibitor or peptide concentration, and the unit of inhibitory activity. The categorization of the relative scale of inhibitory activities among legume samples becomes difficult because of these method differences. Peptide sequences in samples were identified by means of HPLC/MS. Software and online tools were used in bioactivity prediction and computational modelling. The identification of the types and locations of chemical interactions between the inhibitor peptides and enzymes and the type of enzyme inhibition were achieved through computational modelling and enzyme kinetic studies.
Collapse
|
42
|
Saikia J, Washmin N, Borah T, Sarmah P, Konwar P, Siga A, Haldar S, Banik D. Physicochemical properties, chemical composition and sensory attributes of Alpinia nigra (Gaertn.) B.L. Burtt rhizome: an underutilized spice source. Eur Food Res Technol 2023. [DOI: 10.1007/s00217-023-04200-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
43
|
Firdaus JU, Siddiqui N, Alam O, Manaithiya A, Chandra K. Pyrazole scaffold-based derivatives: A glimpse of α-glucosidase inhibitory activity, SAR, and route of synthesis. Arch Pharm (Weinheim) 2023; 356:e2200421. [PMID: 36617511 DOI: 10.1002/ardp.202200421] [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: 08/08/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 01/10/2023]
Abstract
The α-glucosidase is a validated target to develop drugs for treating type 2 diabetes mellitus. The existing α-glucosidase inhibitors have certain shortcomings related to side effects and route of synthesis. Accordingly, it is inevitable to develop new chemical templates as α-glucosidase inhibitors. Pyrazole derivatives have a special place in medicinal chemistry because of various biological activities. Recently, pyrazole-based heterocyclic compounds have emerged as a promising scaffold to develop α-glucosidase inhibitors. This study focuses on the recently reported pyrazole-based α-glucosidase inhibitors, including their biological activity (in vivo, in vitro, and in silico), structure-activity relationship, and ways of synthesis. The literature revealed the development of several promising pyrazole-based α-glucosidase inhibitors and new synthetic routes for their preparation. The encouraging α-glucosidase inhibitory results of the pyrazole-based heterocyclic compounds make them an attractive target for further research. The authors also foresee the arrival of the pyrazole-based α-glucosidase inhibitors in clinical practice.
Collapse
Affiliation(s)
- Jannat Ul Firdaus
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nadeem Siddiqui
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Ozair Alam
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Ajay Manaithiya
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Kailash Chandra
- Department of Biochemistry, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
44
|
In vitro and in-silico inhibitory validation of Tapinanthus cordifolius leaf extract on alpha-amylase in the management of type 2 diabetes. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
45
|
Sharma N, Gupta N, Orfali R, Kumar V, Patel CN, Peng J, Perveen S. Evaluation of the Antifungal, Antioxidant, and Anti-Diabetic Potential of the Essential Oil of Curcuma longa Leaves from the North-Western Himalayas by In Vitro and In Silico Analysis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227664. [PMID: 36431765 PMCID: PMC9695312 DOI: 10.3390/molecules27227664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
Essential oils (EOs) have gained immense popularity due to considerable interest in the health, food, and pharmaceutical industries. The present study aimed to evaluate the antimicrobial and antioxidant activity and the anti-diabetic potential of Curcuma longa leaf (CLO) essential oil. Further, major phytocompounds of CLO were analyzed for their in-silico interactions with antifungal, antioxidant, and anti-diabetic proteins. CLO was found to have a strong antifungal activity against the tested Candida species with zone of inhibition (ZOI)-11.5 ± 0.71 mm to 13 ± 1.41 mm and minimum inhibitory concentration (MIC) was 0.63%. CLO also showed antioxidant activity, with IC50 values of 5.85 ± 1.61 µg/mL using 2,2-diphenyl-1-picrylhydrazyl (DPPH) scavenging assay and 32.92 ± 0.64 µM using ferric reducing antioxidant power (FRAP) assay. CLO also showed anti-diabetic activity with an IC50 of 43.06 ± 1.24 µg/mL as compared to metformin (half maximal inhibitory concentration, IC50-16.503 ± 0.66 µg/mL). Gas chromatography-mass spectrometry (GC-MS) analysis of CLO showed the presence of (-)-zingiberene (17.84%); 3,7-cyclodecadien-1-one, 3,7-dimethyl-10-(1-methylethylidene)-(15.31%); cyclohexene, 4-methyl-3-(1-methylethylidene) (12.47%); and (+)-4-Carene (11.89%) as major phytocompounds. Molecular docking of these compounds with antifungal proteins (cytochrome P450 14 alpha-sterol demethylase, PDB ID: 1EA1, and N-myristoyl transferase, PDB ID: 1IYL), antioxidant (human peroxiredoxin 5, PDB ID: 1HD2), and anti-diabetic proteins (human pancreatic alpha-amylase, PDB ID: 1HNY) showed strong binding of 3,7-cyclodecadien-1-one with all the selected protein targets. Furthermore, molecular dynamics (MD) simulations for a 100 ns time scale revealed that most of the key contacts of target proteins were retained throughout the simulation trajectories. Binding free energy calculations using molecular mechanics generalized born surface area (MM/GBSA), and drug-likeness and toxicity analysis also proved the potential for 3,7-cyclodecadien-1-one, 3,7-dimethyl-10-(1-methylethylidene) to replace toxic synthetic drugs and act as natural antioxidants.
Collapse
Affiliation(s)
- Nitin Sharma
- Department of Biotechnology, Chandigarh College of Technology, CGC, Landran, Mohali 140307, India
- Correspondence: (N.S.); (S.P.)
| | - Nidhi Gupta
- Department of Biotechnology, Chandigarh College of Technology, CGC, Landran, Mohali 140307, India
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Vikas Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India
| | - Chirag N. Patel
- Department of Botany, Bioinformatics, and Climatic Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Jiangnan Peng
- Department of Medicinal, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Shagufta Perveen
- Department of Medicinal, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
- Correspondence: (N.S.); (S.P.)
| |
Collapse
|
46
|
Guo S, Meng J, Wang S, Gu D, Chu T, Yang Y. Preparation of magnetic microcapsules of α-amylase and α-glucosidase for dual-target affinity screening of active components from Toona sinensis. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Chemical Composition, Antibacterial, Antifungal and Antidiabetic Activities of Ethanolic Extracts of Opuntia dillenii Fruits Collected from Morocco. J FOOD QUALITY 2022. [DOI: 10.1155/2022/9471239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Opuntia dillenii (Ker Gawl.) Haw. belongs to the Cactaceae family and is native to the arid and semi-arid regions of Mexico and the southern United States. O. dillenii are now used as medicinal plants in various countries. In this study, we investigated the chemical composition of ethanolic extracts obtained from seeds, juice, and peel of O. dillenii fruits collected from Morocco, and we evaluated their antibacterial, antifungal, and antidiabetic activities. Phytochemical screening revealed high quantities of polyphenols (193.73 ± 81.44 to 341.12 ± 78.90 gallic acid eq [g/100 g dry weight]) in the extracts. The major phenolic compounds determined by HPLC were gallic acid, vanillic acid, and syringic acid. Regarding flavonoids, quercetin 3-O-β-D-glucoside and kaempferol were the predominant molecules. Juice extracts showed weak to moderate antibacterial activity against the bacteria species Listeria monocytogenes, Escherichia coli, and Salmonella braenderup. All tested extracts displayed a significant inhibitory effect on α-glucosidase and α-amylase activities in vitro, with the peel extracts showing the greatest inhibitory effects. Together, these findings suggest that O. dillenii fruits are a promising source for the isolation of novel compounds with antibacterial or antidiabetic activities. For the most abundant phytochemicals identified in O. dillenii peel ethanolic extract, molecular docking simulations against human pancreatic α-amylase enzyme were performed. These indicated the presence of bioactive compounds in the extract with a better potential to decrease the enzyme activity than the commercial drug acarbose.
Collapse
|
48
|
An in vitro study of the 5-methyl- and 5-bromo/chloro substituted 2-hydroxy-3-nitrochalcones as α-glucosidase and/or α-amylase inhibitors with potential anti-inflammatory activity. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
49
|
Markússon S, Hjörleifsson JG, Kursula P, Ásgeirsson B. Structural Characterization of Functionally Important Chloride Binding Sites in the Marine Vibrio Alkaline Phosphatase. Biochemistry 2022; 61:2248-2260. [PMID: 36194497 DOI: 10.1021/acs.biochem.2c00438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Enzyme stability and function can be affected by various environmental factors, such as temperature, pH, and ionic strength. Enzymes that are located outside the relatively unchanging environment of the cytosol, such as those residing in the periplasmic space of bacteria or extracellularly secreted, are challenged by more fluctuations in the aqueous medium. Bacterial alkaline phosphatases (APs) are generally affected by ionic strength of the medium, but this varies substantially between species. An AP from the marine bacterium Vibrio splendidus (VAP) shows complex pH-dependent activation and stabilization in the 0-1.0 M range of halogen salts and has been hypothesized to specifically bind chloride anions. Here, using X-ray crystallography and anomalous scattering, we have located two chloride binding sites in the structure of VAP, one in the active site and another one at a peripheral site. Further characterization of the binding sites using site-directed mutagenesis and small-angle X-ray scattering showed that upon binding of chloride to the peripheral site, structural dynamics decreased locally, resulting in thermal stabilization of the VAP active conformation. Binding of the chloride ion in the active site did not displace the bound inorganic phosphate product, but it may promote product release by facilitating rotational stabilization of the substrate-binding Arg129. Overall, these results reveal the complex nature and dynamics of chloride binding to enzymes through long-range modulation of electronic potential in the vicinity of the active site, resulting in increased catalytic efficiency and stability.
Collapse
Affiliation(s)
- Sigurbjörn Markússon
- Science Institute, University of Iceland, 107 Reykjavik, Iceland.,Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | | | - Petri Kursula
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90570 Oulu, Finland.,Biocenter Oulu, University of Oulu, 90570 Oulu, Finland
| | | |
Collapse
|
50
|
Chen L, Lv B, Zhang X, Xu Y, Wu P, Zhou W, Chen XD. The swallowing threshold and starch hydrolysis of cooked rice with different moisture contents for human mastication. Food Res Int 2022; 160:111677. [DOI: 10.1016/j.foodres.2022.111677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/30/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022]
|