1
|
Mohammadi-Khanaposhtani M, Bahrami T, Bandarian F, Nasli-Esfahani E, Roostaei D, Zamani E, Aghajani F, Mahdavi M, Ahangar N. In vivo anti-hyperglycemic activity and toxicity evaluation of two bis-coumarin derivative as potential α-glucosidase inhibitors. J Diabetes Metab Disord 2025; 24:67. [PMID: 39959578 PMCID: PMC11822155 DOI: 10.1007/s40200-025-01573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/26/2025] [Indexed: 02/18/2025]
Abstract
Objectives The in vivo assay is a key step in the development of a new compound as a drug. In the present work, bis-4-aminocoumarin derivative 3,3'-(p-tolylmethylene)bis(4-amino-2H-chromen-2-one) (PTBAC) and bis-4-hydroxycoumarin derivative 3,3'-((4-((1-(2-chlorobenzyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)methylene)bis(4-hydroxy-2H-chromen-2-one) (2CTMBHC) that showed high anti-α-glucosidase activity on the yeast form of this enzyme were selected for in vivo anti-hyperglycemic assay. Methods The in vivo anti-hyperglycemic effect of PTBAC and 2CTMBHC was assessed using oral starch tolerance test in streptozotocin-induced diabetic albino mouse model, and the results were compared with acarbose as a representative inhibitor of intestinal α-glucosidase enzyme. Toxicity of the selected compounds was also evaluated in vivo. Results The obtained results revealed that both selected compounds, PTBAC and 2CTMBHC, showed more anti-diabetic effects when compared with acarbose as a standard drug. In vivo anti-diabetic assays also demonstrated that bis-4-hydroxycoumarin derivative 2CTMBHC was more potent than bis-4-aminocoumarin derivative PTBAC. In vivo results were also confirmed by in vitro and in silico studies. Moreover, there was not any apparent signs of toxicity and mortality in in vivo toxicity assay. Conclusions In summary, in vivo anti-hyperglycemic effects of two synthetic compounds PTBAC and 2CTMBHC was confirmed in this study. Given that these compounds exhibited no evidences of toxicity and mortality in mice, therefore, they are good candidates for further investigations. Graphical Abstract
Collapse
Affiliation(s)
- Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Bahrami
- Department of Pharmacology-Toxicology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Bandarian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Davoud Roostaei
- Department of Pharmacology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Zamani
- Department of Pharmacology-Toxicology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Forough Aghajani
- Department of Pharmacology-Toxicology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nematollah Ahangar
- Department of Pharmacology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
2
|
Mehreen S, Ali MI, Muntha ST, Zia M, Ullah A, Ullah S, Khan A, Hussain J, Anwar MU, Al-Harrasi A, Naseer MM. Synthesis, structural insights and bio-evaluation of N-phenoxyethylisatin hydrazones as potent α-glucosidase inhibitors. RSC Adv 2025; 15:14717-14729. [PMID: 40337235 PMCID: PMC12056728 DOI: 10.1039/d5ra00770d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 05/01/2025] [Indexed: 05/09/2025] Open
Abstract
Effective α-glucosidase inhibitors are vital for managing type 2 diabetes, emphasizing the need for novel and potent compounds. A series of novel N-phenoxyethylisatin hydrazones 1(a-l) have been synthesized and characterized by their spectral data, and in the case of 1l by its single crystal X-ray analysis. All the synthesized compounds were in vitro evaluated for their inhibition potential against the α-glucosidase enzyme. Interestingly, most of these compounds exhibited significant inhibitory activity against the α-glucosidase enzyme, with IC50 values ranging from 3.64 ± 0.13 to 94.89 ± 0.64 μM compared to the standard drug, acarbose (IC50 = 873.34 ± 1.67 μM). The compound 1e was found to be the most active compound of the series having an IC50 value of 3.64 ± 0.13 μM. Molecular docking studies revealed a binding score of -9.7 kcal mol-1 for 1e, slightly surpassing that of acarbose (-9.4 kcal mol-1). Unlike acarbose, which primarily relies on hydrogen bonding, the binding interactions of 1e are dominated by π-interactions. ADMET profiling confirmed favourable pharmacokinetics for these compounds, including good oral bioavailability, balanced hydrophilicity, and minimal predicted toxicity. These findings highlight the potential of these compounds as promising candidates for the development of more effective treatments for hyperglycemia.
Collapse
Affiliation(s)
- Saba Mehreen
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
- Department of Chemistry, Faculty of Sciences, The University of Haripur KP 22620 Pakistan
| | - Muhammad Imran Ali
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Sidra Tul Muntha
- Peking University Institute of Advanced Agriculture Sciences Weifang Shandong China
| | - Mehwash Zia
- Department of Chemistry, Allama Iqbal Open University Islamabad 44000 Pakistan
| | - Aman Ullah
- Department of Agricultural, Food, and Nutritional Science, 4-10 Agriculture/Forestry Centre, University of Alberta Edmonton AB T6G 2P5 Canada
| | - Saeed Ullah
- Natural and Medical Sciences Research Centre, University of Nizwa Birkat Almouz 616 Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa Birkat Almouz 616 Oman
- Department of Chemical and Biological Engineering, College of Engineering, Korea University Seoul 02841 Republic of Korea
| | - Javid Hussain
- Department of Biological Sciences & Chemistry, College of Arts and Sciences, University of Nizwa Nizwa Oman
| | - Muhammad U Anwar
- Natural and Medical Sciences Research Centre, University of Nizwa Birkat Almouz 616 Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa Birkat Almouz 616 Oman
| | | |
Collapse
|
3
|
Ma X, Li H, Li Y, Xie X, Wang Y, Wang M, Peng X. Potential Antidiabetic Activity of Nordihydroguaiaretic Acid: An Insight into Its Inhibitory Mechanisms on Carbohydrate-Hydrolyzing Enzymes, the Binding Behaviors with Enzymes, and In Vivo Antihyperglycemic Effect. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8290-8304. [PMID: 40152424 DOI: 10.1021/acs.jafc.4c11307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The inhibitory mechanisms and binding behaviors of nordihydroguaiaretic acid (NDGA) to α-glucosidase/α-amylase were investigated by in vitro multispectroscopic methods and in silico modeling technique. The results demonstrated that NDGA reversibly and uncompetitively inhibited α-glucosidase, exhibiting stronger inhibition than acarbose, while it displayed noncompetitive inhibition against α-amylase. Additionally, NDGA could spontaneously bind to α-glucosidase/α-amylase mainly through hydrogen bonds and hydrophobic forces, thus altering the spatial structure of enzymes and reducing their catalytic activity. The presence of crowding reagents/polysaccharides/undigested milk proteins would decrease the inhibitory ability of NDGA, whereas fatty acids exhibited the opposite phenomenon on α-glucosidase. Furthermore, the antidiabetic activity of NDGA in vivo was evaluated using the diabetic Drosophila model induced by a high-sugar diet. It was found that NDGA significantly reduced the glucose levels of diabetic Drosophila. These findings suggested that NDGA was a potential inhibitor of α-glucosidase/α-amylase and could be used as a nutritional adjuvant to prevent diabetes.
Collapse
Affiliation(s)
- Xiangzhao Ma
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Huan Li
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Mengfan Wang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xin Peng
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
4
|
Cai X, Hu S, Lin C, Wu J, Wang J, Wang Z, Zhang X, Wang X, Xu F, Chen L, Yang W, Nie L, Ji L. Comparison of glucose fluctuation between metformin combined with acarbose or sitagliptin in Chinese patients with type 2 diabetes: A multicenter, randomized, active-controlled, open-label, parallel design clinical trial. Chin Med J (Engl) 2025:00029330-990000000-01503. [PMID: 40178116 DOI: 10.1097/cm9.0000000000003477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Alpha-glucosidase inhibitors or dipeptidyl peptidase-4 inhibitors are both hypoglycemia agents that specifically impact on postprandial hyperglycemia. We compared the effects of acarbose and sitagliptin add on to metformin on time in range (TIR) and glycemic variability (GV) in Chinese patients with type 2 diabetes mellitus through continuous glucose monitoring (CGM). METHODS In this multicenter, randomized, open-label, active-controlled study, we recruited patients with type 2 diabetes mellitus aged 18-65 years with body mass index (BMI) within 19-40 kg/m2 and hemoglobin A1c (HbA1c) between 6.5% and 9.0%. Eligible patients were randomized to receive either metformin combined with acarbose 100 mg three times daily or metformin combined with sitagliptin 100 mg once daily for 28 days. After the first 14-day treatment period, patients wore CGM and entered another 14-day treatment period. The primary outcome was the level of TIR after treatment between groups. We also performed time series decomposition, dimensionality reduction, and clustering using the CGM data. RESULTS A total of 701 participants received either acarbose or sitagliptin treatment in combination with metformin. There was no statistically significant difference in TIR between the two groups. Time below range (TBR) and coefficient of variation (CV) levels in acarbose users were significantly lower than those in sitagliptin users. TBR below target level <3.9 mmol/L (TBR3.9): Acarbose: 2.86 ± 6.98% vs. Sitagliptin: 3.89 ± 9.43%, P = 0.042; TBR3.0: Acarbose: 0.96 ± 4.41% vs. Sitagliptin: 1.64 ± 6.73%, P = 0.033; CV: Acarbose: 22.44 ± 5.08% vs. Sitagliptin: 23.96 ± 5.19%, P <0.001. No significant difference was found in the complexity of glucose time series index (CGI) between acarbose users and sitagliptin users. By using time series analysis and clustering, we distinguished three groups of patients with representative metabolism characteristics, especially in GV. CONCLUSIONS Acarbose had slight advantages over sitagliptin in improving GV and reducing the risk of hypoglycemia. Time series analysis of CGM data may predict GV and the risk of hypoglycemia. TRIAL REGISTRATION Chinese Clinical Trial Registry: ChiCTR2000039424.
Collapse
Affiliation(s)
- Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Suiyuan Hu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Jing Wu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Junfen Wang
- Department of Endocrinology and Metabolism, The Second Hospital of Shijiazhuang, Shijiazhuang, Hebei 050000, China
| | - Zhufeng Wang
- Department of Endocrinology and Metabolism, Guang'anmen Hospital, China Academy of Chinese Medical Sciences (South Area), Beijing 102600, China
| | - Xiaomei Zhang
- Department of Endocrinology and Metabolism, Peking University International Hospital, Beijing 102206, China
| | - Xirui Wang
- Department of Endocrinology and Metabolism, Beijing Airport Hospital, Beijing 101300, China
| | - Fengmei Xu
- Department of Endocrinology and Metabolism, Hebi Coal (group) Ltd. General Hospital, Hebi, Henan 458030, China
| | - Ling Chen
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Wenjia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Lin Nie
- Department of Endocrinology and Metabolism, Beijing Airport Hospital, Beijing 101300, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
5
|
Abbasi SA, Rahim F, Hussain R, Rehman W, Khan S, Taha M, Iqbal T, Khan Y, Ali Shah SA. Synthesis of modified Schiff base appended 1,2,4-triazole hybrids scaffolds: elucidating the in vitro and in silico α-amylase and α-glucosidase inhibitors potential. Z NATURFORSCH C 2025; 80:119-134. [PMID: 38996406 DOI: 10.1515/znc-2024-0073] [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: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024]
Abstract
The current study involves the synthesis of Schiff bases based on 1,2,4-triazoles skeleton and assessing their α-amylase and α-glucosidase profile. Furthermore, the precise structures of the synthesized derivatives were elucidated using various spectroscopic methods such as 1H-NMR, 13C-NMR and HREI-MS. Using glimepiride as the reference standard, the in vitro α-glucosidase and α-amylase inhibitory activities of the synthesized compounds were evaluated in order to determine their potential anti-diabetic properties. All analogues showed varied range of inhibitory activity having IC50 values ranging from 17.09 ± 0.72 to 45.34 ± 0.03 μM (α-amylase) and 16.35 ± 0.42 to 42.31 ± 0.09 μM (α-glucosidase), respectively. Specifically, the compounds 1, 7 and 8 were found to be significantly active with IC50 values of 17.09 ± 0.72, 19.73 ± 0.42, and 23.01 ± 0.04 μM (against α-amylase) and 16.35 ± 0.42, 18.55 ± 0.26, and 20.07 ± 0.02 μM (against α-glucosidase) respectively. The obtained results were compared with the Glimepiride reference drug having IC50 values of 13.02 ± 0.11 μM (for α-glucosidase) and 15.04 ± 0.02 μM (for α-amylase), respectively. The structure-activity relationship (SAR) studies were conducted based on differences in substituent patterns at varying position of aryl rings A and B may cause to alter the inhibitory activities of both α-amylase and α-glucosidase enzymes. Additionally, the molecular docking study was carried out to explore the binding interactions possessed by most active analogues with the active sites of targeted α-amylase and α-glucosidase enzymes.
Collapse
Affiliation(s)
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Rafaqat Hussain
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Wajid Rehman
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Shoaib Khan
- Department of Chemistry, Abbottabad University of Science and Technology (AUST), Abbottabad, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982 31441, Dammam, Saudi Arabia
| | - Tayyiaba Iqbal
- Department of Chemistry, Abbottabad University of Science and Technology (AUST), Abbottabad, Pakistan
| | - Yousaf Khan
- Department of Chemistry, COMSATS University Islamabad, 45550, Islamabad, Pakistan
| | - 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 Products Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia
| |
Collapse
|
6
|
Yu Y, Wang H, Jin X, Huang W, Zhao Y, Wang N, Lu D, Wei B, Wang H. Structural Characterization of Dendrobium officinale Polysaccharides and Their Regulation Effect on Intestinal Microbiota During In Vitro Fermentation. Polymers (Basel) 2025; 17:727. [PMID: 40292547 PMCID: PMC11944827 DOI: 10.3390/polym17060727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 04/30/2025] Open
Abstract
Polysaccharides derived from Dendrobium officinale have been demonstrated to exhibit metabolic regulatory properties. However, the correlation between their structure and function, particularly their mechanism of action through gut microbiota, remains underexplored. This study systematically elucidates the structural characteristics of Dendrobium officinale polysaccharide (DOP) from the Guizhou (GZ) and Zhejiang (ZJ) provinces of China using nuclear magnetic resonance (NMR) and a series of chromatographic analyses, revealing their unique molecular features. Additionally, the metabolic regulatory activities were assessed through α-glucosidase inhibitory assay and in vitro intestinal flora activity assay. The findings include the following: (1) both DOP-GZ and DOP-ZJ predominantly consist of glycosidic linkages of β-1,4-Manp and β-1,4-Glcp; (2) zhe monosaccharide composition ratios of mannose to glucose are 2.51:1 for DOP-GZ and 2.66:1 for DOP-ZJ, with molecular weights of 356 kDa and 544 kDa, respectively, indicating significant structural differences between DOPs from different sources; (3) treatment with DOP-GZ and DOP-ZJ led to alterations in the α-diversity indices and Firmicutes-to-Bacteroidota ratios; (4) more importantly, DOP-GZ and DOP-ZJ significantly increase the abundance of beneficial bacteria (e.g., g_Proteobacteria_unclassified) while suppressing the growth of pathogenic bacteria (e.g., f_Enterobacteriaceae_unclassified), with statistically significant results. These findings not only uncover a novel mechanism by which DOPs regulate metabolism through gut microbiota but also provide a crucial theoretical basis for the application of DOPs in functional foods and pharmaceutical development.
Collapse
Affiliation(s)
- Yanlei Yu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Honggang Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Xiaoshu Jin
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Wenjing Huang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Yunjie Zhao
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Ningning Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Dongze Lu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
- Binjiang Cyberspace Security Institute of ZJUT, Hangzhou 310056, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
- Binjiang Cyberspace Security Institute of ZJUT, Hangzhou 310056, China
| |
Collapse
|
7
|
Mengyuan Z, Chen C, Feng W, Ning Z, Wanyu Y, Tianrong Z, Guoyan R, Zhijun Q, Bin Z. Identification and Molecular Mechanism of Novel α-Glucosidase Inhibitory Peptides from the Hydrolysate of Hemp Seed Proteins: Peptidomic Analysis, Molecular Docking, and Dynamics Simulation. Int J Mol Sci 2025; 26:2222. [PMID: 40076843 PMCID: PMC11899805 DOI: 10.3390/ijms26052222] [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: 01/27/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
There is a growing demand for natural and potent α-glucosidase inhibitors due to the rising prevalence of diabetes. In this study, newly identified α-glucosidase inhibitory peptides were identified from the tryptic hydrolysate of hemp seed proteins based on peptidomics and in silico analysis. A total of 424 peptides, primarily derived from four cupin-type-1 domain-containing proteins, were identified, and 13 ultimately were selected for validation based on their higher PeptideRanker scores, solubility, non-toxicity, and favorable ADMET properties. Molecular docking revealed that these 13 peptides primarily interacted with α-glucosidase via hydrogen bonding and hydrophobic interactions. Among them, three novel peptides-NPVSLPGR (-8.7 kcal/mol), LSAERGFLY (-8.5 kcal/mol), and PDDVLANAF (-8.4 kcal/mol)-demonstrated potent α-glucosidase inhibitory activity due to their lower binding energies than acarbose (-8.1 kcal/mol), the first approved α-glucosidase inhibitor for type 2 diabetes treatment. The molecular mechanism analysis revealed that the peptides NPVSLPGR and LSAERGFLY inhibited α-glucosidase by simultaneously blocking substrate entry through occupying the entrance of the active site gorge and preventing catalysis by binding to active sites. In contrast, the peptide PDDVLANAF primarily exerted inhibitory effects by occupying the entrance of the active site gorge. Molecular dynamics simulation validated the stability of the complexes and provided additional insights into the molecular mechanism determined through docking. These findings contribute essential knowledge for the advancement of natural α-glucosidase inhibitors and offer a promising approach to effectively manage diabetes.
Collapse
Affiliation(s)
- Zhang Mengyuan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Chen Chen
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Wei Feng
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Zhao Ning
- Academy of Military Medical Sciences, Beijing 100850, China;
| | - Yang Wanyu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Zhang Tianrong
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Ren Guoyan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Qiu Zhijun
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
| | - Zhang Bin
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (Z.M.); (C.C.); (W.F.); (Y.W.); (Z.T.); (R.G.); (Q.Z.)
- Henan Engineering Research Center of Food Microbiology, Luoyang 471023, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang 471023, China
| |
Collapse
|
8
|
Waseem T, Zargaham MK, Ahmed M, Rajput TA, Amin A, Nadeem H. Computational investigation to identify multi-targeted anti-hyperglycemic potential of substituted 2-Mercaptobenzimidazole derivatives and synthesis of new α-glucosidase inhibitors. J Comput Aided Mol Des 2025; 39:9. [PMID: 39992499 DOI: 10.1007/s10822-025-00587-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
One of the most widespread diseases recognized all over the world is diabetes, accounting for 1.5 million deaths each year. Recent studies have demonstrated benzimidazole derivatives as potential antidiabetic agents. Hence, the present study is focused on designing new derivatives of 2-mercaptobenzimidazole by C-S cross-coupling reaction and are subjected to computational screening to identify the most promising candidate. Molecular docking and MM-GBSA calculations were performed to ascertain the binding potential with different antidiabetic targets, including α-glucosidase, PPaR-γ, DPP-4, and AMPK. We observed somewhat moderate binding interactions of the synthesized compound against the α-glucosidase. Since binding affinities can be improved using synthetic chemistry approaches, synthesis of analogues (A-18a-c) by designing hybrids at sites such as the acidic functionality of A-18 was done. The analogue A-18a, with p-fluorobenzyl substitution, exhibited enhanced binding affinity (-4.339 Kcal/mol) with the α-glucosidase compared to the parent compound (-3.827 Kcal/mol). The synthesized analogues were also subjected to an in-vitro α-glucosidase inhibitory assay. Among them, A-18a exhibited the most significant inhibitory potential, with an IC50 value of 0.521 ± 0.01 µM as compared to the standard drug Acarbose (IC50 21.0 ± 0.5 µM). This aligns with the computational study findings, where A-18a exhibited stronger binding interactions within the active site of the enzyme. Hence, a promising analogue of the designed compound was synthesized through a computationally guided approach as an anti-hyperglycaemic agent. Additionally, most of the designed compounds showed significantly greater binding affinity with PPaR-γ as compared to the standard pioglitazone. A-18 was successfully synthesized by S-arylation reaction using CuI in 89% yield and was subjected to MD-simulation against PPaR-γ, which revealed stable binding throughout the 200 ns run. Future studies will focus on exploring the activity of the designed drugs against PPaR-γ through in-vitro and in-vivo assays.
Collapse
Affiliation(s)
- Tanya Waseem
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Muhammad Kazim Zargaham
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Madiha Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan.
| | - Tausif Ahmed Rajput
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Adnan Amin
- NPRL Department of Pharmacognosy, Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| |
Collapse
|
9
|
Kim MH, Yeon SW, Ryu SH, Lee HH, Turk A, Jeong SY, Kim YJ, Lee KY, Hwang BY, Lee MK. Structural Diversity and Anti-Diabetic Potential of Flavonoids and Phenolic Compounds in Eriobotrya japonica Leaves. Molecules 2025; 30:736. [PMID: 39942838 PMCID: PMC11820478 DOI: 10.3390/molecules30030736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Eriobotrya japonica (Thunb.) Lindl., commonly known as loquat, is a plant belonging to the Rosaceae family. While its fruit is widely consumed as food and used in traditional medicine, research on other parts of the plant remains insufficient. Therefore, the chemical constituents and biological activities of its leaves were investigated. Phytochemical analysis of E. japonica leaves identified 30 compounds, including flavonoids, phenolics, and megastigmanes. The flavonoids isolated from the leaves include flavones, flavans, flavolignans, flavonoid glycosides, and coumaroyl flavonoid glycosides. Coumaroyl flavonoid rhamnosides were characteristically present in E. japonica leaves, and the configurations of coumaric acids, as well as the binding position to the rhamnose in each compound, were identified through detailed NMR analysis. Notably, three of them were isolated from this plant for the first time. Phenolic compounds were found to be present as conjugates with organic acids, such as quinic acid, shikimic acid, and glucose. Flavonoid and phenolic compounds demonstrated significant antioxidant and α-glucosidase inhibitory effects, whereas megastigmanes showed little activity. Notably, coumaroyl flavonoid rhamnosides, which consist of flavonoids combined with the phenolic acid, coumaric acid, exhibited excellent anti-diabetic effects. Further molecular docking analysis confirmed that these compounds effectively bind to the α-glucosidase enzyme. In conclusion, the present study identified flavonoid and phenolic components with various structures in E. japonica leaves and clarified their anti-diabetic and antioxidant effects. These findings support the beneficial potential of E. japonica leaves for the treatment and/or prevention of metabolic diseases.
Collapse
Affiliation(s)
- Min Hee Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - Sang Won Yeon
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - Se Hwan Ryu
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - Hak Hyun Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - Ayman Turk
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - So Yeong Jeong
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - Young Jun Kim
- College of Pharmacy, Korea University, Sejong 47236, Republic of Korea; (Y.J.K.); (K.Y.L.)
| | - Ki Yong Lee
- College of Pharmacy, Korea University, Sejong 47236, Republic of Korea; (Y.J.K.); (K.Y.L.)
| | - Bang Yeon Hwang
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| | - Mi Kyeong Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (M.H.K.); (S.W.Y.); (S.H.R.); (H.H.L.); (A.T.); (S.Y.J.); (B.Y.H.)
| |
Collapse
|
10
|
Choudhry SS, Mehmood H, Akhtar T, Haroon M, Musa M, Sajid Z. Design, Synthesis, and In Silico Molecular Docking Studies of Adamantanyl Hydrazinylthiazoles as Potential Antidiabetic Agents. Chem Biodivers 2025:e202402409. [PMID: 39856497 DOI: 10.1002/cbdv.202402409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 01/27/2025]
Abstract
Diabetes mellitus (DM) is a widespread disease that poses a major threat to millions of people. To address this issue, we have synthesized seventeen new 4-(adamantan-1-yl)-(2-(arylidene)hydrazinyl)thiazoles (3a-q) via Hantzsch synthetic approach. The molecular structures of all the compounds were confirmed using FT-IR, 1H- and 13C-NMR spectroscopy, and HR-mass spectrometry. Protein kinase, α-amylase, glycation, and oxidation inhibition potential of all compounds were also investigated, and it was found that compounds 3b, 3c, 3e-3g, and 3i-3q have shown excellent α-amylase inhibition (IC50 = 7.91 ± 0.07 to 28.57 ± 0.1 µM), compounds 3c, 3e, 3i, 3k, and 3p (IC50 = 30.6 ± 0.06 to 37.8 ± 0.005 ppm) were found to be highly potent anti-glycating agents, and compounds 3c, 3g, 3h, 3k, and 3m were found to be more potent protein kinase inhibitors as compared to standards. The compounds 3b, 3c, 3d, 3e, 3f, 3g, 3i, 3k, 3l, 3m, 3n, 3p, and 3q have shown good antioxidant potential (IC50 = 27.5 ± 0.09 to 48.8 ± 0.09 µM) as compared to standard ascorbic acid (IC50 = 51.3 ± 0.1 µM). The biocompatibility of all samples was also tested by employing brine shrimp lethality and in vitro hemolytic assays and was found to be safe to human erythrocytes at tested concentrations. Furthermore, the molecular docking simulation study also revealed that almost all synthesized compounds have potential interactions with target proteins at the molecular level.
Collapse
Affiliation(s)
- Sabah Siddique Choudhry
- Department of Chemistry, Mirpur University of Science and Technology (MUST), Mirpur, Azad Kashmir, Pakistan
| | - Hasnain Mehmood
- Department of Chemistry, Mirpur University of Science and Technology (MUST), Mirpur, Azad Kashmir, Pakistan
| | - Tashfeen Akhtar
- Department of Chemistry, Mirpur University of Science and Technology (MUST), Mirpur, Azad Kashmir, Pakistan
| | - Muhammad Haroon
- Department of Chemistry and Biochemistry, Miami University, 651 E High St, Oxford, Ohio, United States
| | - Mustapha Musa
- GSK Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Nottingham, UK
| | - Zaroon Sajid
- Department of Chemistry, Mirpur University of Science and Technology (MUST), Mirpur, Azad Kashmir, Pakistan
| |
Collapse
|
11
|
Wang C, Li YP, Gong X, Gan LS, Zhang H. Rare diphenylheptanoid-phenylheptanoid hybrids with α-glucosidase inhibitory effects from the pollen of Typha angustifolia. Nat Prod Res 2025; 39:20-30. [PMID: 37599620 DOI: 10.1080/14786419.2023.2248352] [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: 05/18/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 08/22/2023]
Abstract
Two rarely occurring diphenylheptanoid-phenylheptanoid hybrid dimers (1 and 2) and one new oxygenated fatty acid (3), as well as two known fatty acid analogues (4 and 5), were isolated from the 70% EtOH extract of the pollen of Typha angustifolia. Their planar structures were established by interpretation of MS and NMR spectroscopic data, and the absolute configurations of 1 and 2 were determined by Mosher's method and quantum chemical TD-DFT calculations of ECD spectra. An in vitro anti-diabetic evaluation of these isolates revealed that compounds 1 and 2 exhibited promising inhibitory activity against α-glucosidase with IC50 values of 11.85 ± 0.69 and 17.06 ± 3.08 μM, respectively. It is the first report on both diphenylheptanoid constituents and α-glucosidase inhibitors from the title plant, which represents a significant phytochemical progress of this herbal species and may serve as a reference for its future medicinal applications.
Collapse
Affiliation(s)
- Chao Wang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Yu-Peng Li
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Xu Gong
- School of Biotechnology and Health Sciences, International Healthcare Innovation Institute, Wuyi University, Jiangmen, China
| | - Li-She Gan
- School of Biotechnology and Health Sciences, International Healthcare Innovation Institute, Wuyi University, Jiangmen, China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| |
Collapse
|
12
|
Razon AH, Alauddin M, Farzana N, Mazumdar S, Amin MR, Tusher MMH, Asrafuzzaman M, Hasan N, Rahman M, Saiedullah M, Rokeya B, Faruque MO. The Intricate Mechanisms of Functional Foods Oyster Mushroom and Fenugreek on Type 2 Diabetic Animal Model. J Diabetes Res 2024; 2024:6209785. [PMID: 39885962 PMCID: PMC11779994 DOI: 10.1155/jdr/6209785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/19/2024] [Indexed: 02/01/2025] Open
Abstract
Mushrooms and fenugreek are widely used to reduce hyperglycemia, and fenugreek is also used as a culinary ingredient to enhance flavor and aroma. This study is aimed at investigating the underlying mechanisms of the hypoglycemic effects of mushrooms and fenugreek in a Type 2 diabetic rat model. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) functions to reduce hyperglycemia through insulin-independent pathways and protects beta-cells. Diabetic model rats were administered standard diets supplemented with 5% oyster mushroom powder (mushroom-treated (MT) group) and 5% fenugreek seed powder (fenugreek-treated (FT) group) for 8 weeks. The results showed improvements in both glycemic and lipid profiles, with both oyster mushroom and fenugreek enhancing the phosphorylation of AMPK in muscle tissue. However, no effect on insulin secretion was observed. These findings suggest that both substances reduce hyperglycemia through an insulin-independent pathway. In silico analysis of both mushroom and fenugreek seed extracts revealed bioactive compounds having a strong binding affinity to α-glucosidase, which suggests mushroom and fenugreek supplements might control postprandial blood glucose levels.
Collapse
Affiliation(s)
- Arafat Hassan Razon
- School of Science and Technology, Bangladesh Open University, Gazipur, Bangladesh
| | - Md. Alauddin
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Nisat Farzana
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Sanaullah Mazumdar
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Ruhul Amin
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | | | - Md. Asrafuzzaman
- Department of Pharmacology, Bangladesh University of Health Sciences, Dhaka, Bangladesh
| | - Nahid Hasan
- Department of Biochemistry and Molecular Biology, Bangladesh University of Health Sciences, Dhaka, Bangladesh
| | - Mahfuzur Rahman
- Department of Biochemistry and Molecular Biology, Bangladesh University of Health Sciences, Dhaka, Bangladesh
| | - Muhammad Saiedullah
- Department of Biochemistry and Molecular Biology, Bangladesh University of Health Sciences, Dhaka, Bangladesh
| | - Begum Rokeya
- Department of Pharmacology, Bangladesh University of Health Sciences, Dhaka, Bangladesh
| | - Md. Omar Faruque
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| |
Collapse
|
13
|
Mohammadi-Khanaposhtani M, Bakhtiari N, Bandarian F, Larijani B, Mahdavi M, Najafzadehvarzi H. In vivo anti-diabetic and anti-lipidemic evaluations of an excellent synthetic α-glucosidase inhibitor with dihydropyrano[3,2-c]quinoline skeleton. J Diabetes Metab Disord 2024; 23:2375-2384. [PMID: 39610500 PMCID: PMC11599671 DOI: 10.1007/s40200-024-01505-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/26/2024] [Indexed: 11/30/2024]
Abstract
Objectives The in vivo assay is a key step in the development of a new bioactive compound as a lead drug structure. Based on importance of α-glucosidase inhibitors in the control of blood glucose level (BGL) in diabetes, in the present work, 3-amino-1-(4-chlorophenyl)-12-oxo-11,12-dihydro-1H-benzo[h]pyrano[3,2-c]quinoline-2-carbonitrile (ACODDHBPQC) that showed excellent inhibitory activity on the yeast form of α-glucosidase was selected for in vivo anti-diabetic assay. Methods The in vivo anti-diabetic and anti-lipidemic effects of this synthetic compound were evaluated using by a streptozotocin (STZ)-induced diabetic Wistar rat model. In silico docking study of ACODDHBPQC was performed by Atodock tools and absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties of this compound was predicted by PreADMT online software. Results The obtained results revealed that selected compound ACODDHBPQC showed a significant anti-diabetic effect on diabetic rats. In vivo anti-lipidemic assay also demonstrated that ACODDHBPQC had favorable effects on cholesterol and LDL levels. Furthermore, in silico studies showed that ACODDHBPQC interacted with key residues of the α-glucosidase active site and had good pharmacokinetic and toxicity properties. Conclusion In summary, anti-hyperglycemic effects of ACODDHBPQC was confirmed by in vivo study. However, more evaluations are needed to introduce ACODDHBPQC as a lead drug compound. Graphical abstract
Collapse
Affiliation(s)
- Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Navid Bakhtiari
- Faculty of Pharmacy, Islamic Azad University, Ayatollah Amoli Branch, Amol, Iran
| | - Fatemeh Bandarian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Najafzadehvarzi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
14
|
Ayan EK, Çoban G, Soyer Z. Design, synthesis, biological evaluation, and molecular modeling studies of some quinazolin-4(3 H)-one-benzenesulfonamide hybrids as potential α-glucosidase inhibitors. J Biomol Struct Dyn 2024:1-21. [PMID: 39539169 DOI: 10.1080/07391102.2024.2427373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/30/2024] [Indexed: 11/16/2024]
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by hyperglycemia, posing serious health risks and becoming increasingly prevalent. Prolonged hyperglycemia can lead to complications such as nephropathy, neuropathy, retinopathy, cardiovascular damage, and blindness. Controlling hyperglycemia through α-glucosidase inhibitors, which slow down carbohydrate breakdown, is an effective treatment strategy. However, current inhibitors like acarbose, voglibose, and miglitol while used to manage type 2 diabetes, have significant side effects. Therefore, developing new α-glucosidase inhibitors that are more effective and have fewer side effects is crucial. In this study, a series of novel quinazolin-4(3H)-one-benzenesulfonamide hybrid compounds were designed, synthesized, and evaluated for in vitro α-glucosidase inhibitory activity. The compounds showed higher enzyme inhibition potency, with IC50 values ranging between 129.2 ± 0.5 and 558.7 ± 13.7 µM, compared to acarbose (IC50=814.3 ± 13.5 µM). Among the tested compounds, compound 10, bearing a 4-chlorophenyl ring on the nitrogen atom of the sulfonamide group, was the most active, with an IC50 value of 129.2 ± 0.5 µM. Enzyme kinetics analyses and molecular modeling studies were conducted to understand their inhibition mechanisms and interactions with the enzyme. The kinetic studies revealed a mixed-type inhibition model, indicating that the compounds bind to the enzyme-substrate complex with higher affinity than to the free enzyme. Molecular modeling results confirmed these findings. Additionally, in silico prediction studies showed that the selected compounds have favourable physicochemical and drug-like properties. These results suggest these compounds have potential for further optimization and development as effective α-glucosidase inhibitors for diabetes treatment.
Collapse
Affiliation(s)
- Emre Kadir Ayan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Süleyman Demirel University, Isparta, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey
| | - Güneş Çoban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey
| | - Zeynep Soyer
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey
| |
Collapse
|
15
|
Fadaly WAA, Elshewy A, Nemr MTM, Abdou K, Sayed AM, Kahk NM. Discovery of novel thiazole derivatives containing pyrazole scaffold as PPAR-γ Agonists, α-Glucosidase, α-Amylase and COX-2 inhibitors; Design, synthesis and in silico study. Bioorg Chem 2024; 152:107760. [PMID: 39197383 DOI: 10.1016/j.bioorg.2024.107760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
A novel series of thiazole derivatives with pyrazole scaffold 16a-l as hybrid rosiglitazone/celecoxib analogs was designed, synthesized and tested for its PPAR-γ activation, α-glucosidase, α-amylase and COX-2 inhibitory activities. Regarding the anti-diabetic activity, all compounds were assessed in vitro against PPAR-γ activation, α-glucosidase and α-amylase inhibition in addition to in vivo hypoglycemic activity (one day and 15 days studies). Compounds 16b, 16c, 16e and 16 k showed good PPAR-γ activation (activation % ≈ 72-79 %) compared to that of the reference drug rosiglitazone (74 %). In addition, the same derivatives 16b, 16c, 16e and 16 k showed the highest inhibitory activities against α-glucosidase (IC50 = 0.158, 0.314, 0.305, 0.128 μM, respectively) and against α-amylase (IC50 = 32.46, 23.21, 7.74, 35.85 μM, respectively) compared to the reference drug acarbose (IC50 = 0.161 and 31.46 μM for α-glucosidase and α-amylase, respectively). The most active derivatives 16b, 16c, 16e and 16 k also revealed good in vivo hypoglycemic effect comparable to that of rosiglitazone. In addition, compounds 16b and 16c had the best COX-2 selectivity index (S.I. = 18.7, 31.7, respectively) compared to celecoxib (S.I. = 10.3). In vivo anti-inflammatory activity of the target derivatives 16b, 16c, 16e and 16 k supported the results of in vitro screening as the derivatives 16b and 16c (ED50 = 8.2 and 24 mg/kg, respectively) were more potent than celecoxib (ED50 = 30 mg/kg). In silico docking, ADME, toxicity, and molecular dynamic studies were carried out to explain the interactions of the most active anti-diabetic and anti-inflammatory compounds 16b, 16c, 16e and 16 k with the target enzymes in addition to their physiochemical parameters.
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ahmed Elshewy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini street 11562, Cairo, Egypt; Department of Natural and Applied Sciences, College of Arts and Sciences, The American University of Iraq-Baghdad (AUIB), Baghdad, Iraq
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini street 11562, Cairo, Egypt.
| | - Kareem Abdou
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Collage of Pharmacy, Almaaqal University, 61014 Basrah, Iraq
| | - Nesma M Kahk
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
16
|
Bi S, Xu Z, Wang Z, Liu Y, Yu B, Tian J, Liu C, Qiao L, Zhang Y. Polydatin from Polygoni Cuspidati Rhizoma et Radix regulates glucolipid metabolism in the liver of diabetic rats: Multiscale analysis of network pharmacology and multiomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155992. [PMID: 39216300 DOI: 10.1016/j.phymed.2024.155992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/11/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Polygoni Cuspidati Rhizoma et Radix (Huzhang in Chinese), refers to the root and rhizome of Polygonum cuspidatum Sieb. et Zucc. Huzhang is commonly used in clinical practice for the prevention and treatment of diabetes and its complications, but its active components and regulatory mechanisms have not yet been thoroughly analyzed. PURPOSE The network pharmacology combined with multi-omics analysis will be employed to dissect the substance basis and action mechanism of Huzhang in exerting its anti-diabetic activity. METHODS This study employed phenotypic indicators for baseline assessment, followed by integrated analysis using network pharmacology, metabolomics, transcriptomics, and qPCR technology to elucidate the active components and pharmacological mechanisms of Huzhang. RESULTS The analysis of network pharmacology revealed that polydatin is a potential active component responsible for the anti-T2DM pharmacological effects of Huzhang. In vivo experimental results demonstrated that polydatin significantly regulates blood glucose, lipid levels, liver function, and liver pathological damage in diabetic rats. Analysis results from transcriptomics, metabolomics, and qPCR validation showed that polydatin comprehensively regulates glucose and lipid metabolism in T2DM by modulating bile acid metabolism, fatty acid oxidation, and lipogenesis. CONCLUSION Polydatin is a key component of Huzhang in treating T2DM, and its regulatory mechanisms are diverse, indicating significant development potential.
Collapse
Affiliation(s)
- Shijie Bi
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhenzhen Xu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zewen Wang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanxia Liu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Bin Yu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaye Tian
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Chaoqun Liu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Liansheng Qiao
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanling Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
17
|
Goron AR, Connolly C, Valdez-Sinon AN, Hesson A, Helou C, Kirschen GW. Anti-Hyperglycemic Medication Management in the Perioperative Setting: A Review and Illustrative Case of an Adverse Effect of GLP-1 Receptor Agonist. J Clin Med 2024; 13:6259. [PMID: 39458209 PMCID: PMC11509032 DOI: 10.3390/jcm13206259] [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: 08/18/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
A host of anti-hyperglycemic agents are currently available and widely prescribed for diabetes and weight loss management. In patients undergoing surgery, use of these agents poses a clinical challenge to surgeons, anesthesiologists, and other perioperative care providers with regard to optimal timing of discontinuation and resumption of use, as well as possible effects of these agents on physiology and risk of postoperative complications. Here, we provide a comprehensive review of anti-hyperglycemic medications' effects on physiology, risks/benefits, and best practice management in the perioperative setting. Additionally, we report an illustrative case of small bowel obstruction in a patient taking semaglutide for 6 months prior to an otherwise uncomplicated laparoscopic hysterectomy and bilateral salpingo-oophorectomy. This review is meant to serve not as a replacement of, but rather as a consolidated complement to, various society guidelines regarding perioperative anti-hyperglycemic agent management.
Collapse
Affiliation(s)
- Abby R. Goron
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Courtney Connolly
- Department of Gynecology and Obstetrics, Johns Hopkins Hospital, Baltimore, MD 21287, USA; (C.C.); (A.N.V.-S.)
| | - Arielle N. Valdez-Sinon
- Department of Gynecology and Obstetrics, Johns Hopkins Hospital, Baltimore, MD 21287, USA; (C.C.); (A.N.V.-S.)
| | - Ashley Hesson
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Christine Helou
- Department of Obstetrics and Gynecology, Greater Baltimore Medical Center, Towson, MD 21204, USA;
| | - Gregory W. Kirschen
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Rouzi K, Mortada S, Hassan M, Alsalme A, Kloczkowski A, Karbane ME, Bouatia M, Faouzi MEA, Karrouchi K. Novel 3,5‐Dimethylpyrazole‐Linked 1,2,4‐Triazole‐3‐thiols as Potent Antihyperglycemic Agents: Synthesis, Biological Evaluation, and In Silico Molecular Modelling Investigations. ChemistrySelect 2024; 9. [DOI: 10.1002/slct.202403661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 01/04/2025]
Abstract
AbstractIn this work, a series of pyrazole‐linked 1,2,4‐triazole‐3‐thiol derivatives (3a–i) were prepared and identified by 13C NMR, 1H NMR, and mass spectrometry (ESI‐MS) data. The newly synthesized molecules were also evaluated in vitro for their α‐amylase and α‐glucosidase inhibitory potential. All newly synthesized compounds exhibited potent α‐glucosidase inhibition activity with IC50 in the range of 1.016 ± 0.70 to 24.40 ± 0.02 µM and good α‐amylase inhibitory with IC50 in the range of 49.91 ± 0.32 to 500 µM, as compared to acarbose. The most potent compound among this series is derivative 3e, with IC50 value of 1.016 ± 0.70 µM, which is many folds more than that of acarbose. In addition, in docking studies, both compounds exhibited good interactions at the active region of target proteins. Therefore, this study may lead via structural modifications to the discovery of new potent α‐amylase and α‐glucosidase inhibitors useful in the diabetes treatment.
Collapse
Affiliation(s)
- Khouloud Rouzi
- Laboratory of Analytical Chemistry and Bromatology Team of Formulation and Quality Control of Health Products Faculty of Medicine and Pharmacy Mohammed V University in Rabat Rabat Morocco
| | - Salma Mortada
- Laboratory of Pharmacology and Toxicology Faculty of Medicine and Pharmacy University Mohammed V in Rabat Rabat Morocco
| | - Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine Nationwide Children's Hospital Columbus Ohio 43205 USA
| | - Ali Alsalme
- Department of Chemistry, College of Science King Saud University Riyadh 11451 Saudi Arabia
| | | | - Miloud El Karbane
- Laboratory of Analytical Chemistry and Bromatology Team of Formulation and Quality Control of Health Products Faculty of Medicine and Pharmacy Mohammed V University in Rabat Rabat Morocco
| | - Mustapha Bouatia
- Laboratory of Analytical Chemistry and Bromatology Team of Formulation and Quality Control of Health Products Faculty of Medicine and Pharmacy Mohammed V University in Rabat Rabat Morocco
| | - My El Abbes Faouzi
- Laboratory of Pharmacology and Toxicology Faculty of Medicine and Pharmacy University Mohammed V in Rabat Rabat Morocco
| | - Khalid Karrouchi
- Laboratory of Analytical Chemistry and Bromatology Team of Formulation and Quality Control of Health Products Faculty of Medicine and Pharmacy Mohammed V University in Rabat Rabat Morocco
| |
Collapse
|
19
|
Babalola BA, Sharma L, Olowokere O, Malik M, Folajimi O. Advancing drug discovery: Thiadiazole derivatives as multifaceted agents in medicinal chemistry and pharmacology. Bioorg Med Chem 2024; 112:117876. [PMID: 39163743 DOI: 10.1016/j.bmc.2024.117876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024]
Abstract
In this dispensation of rapid scientific and technological advancements, significant efforts are being made to curb health-related diseases. Research discoveries have highlighted the value of heterocyclic compounds, particularly thiadiazole derivatives, due to their diverse pharmacological activities. These compounds play a crucial role in therapeutic medicine and the development of effective drugs. Thiadiazoles are five-membered heterocyclic compounds consisting of one sulfur and two nitrogen atoms. This review explores advanced synthesis techniques, including the use of heterogeneous catalysts, microwave-assisted methods, ultrasound-assisted synthesis, solvent-free processes, multicomponent reactions, copper-catalyzed aerobic oxidative annulation, intramolecular cyclization, click-chemistry supported synthesis, and alkali-promoted, transition-metal-free mediated synthesis. These methods enhance the diversity and potential applications of thiadiazole compounds. Furthermore, this study provides up-to-date information on the key pharmacological activities of thiadiazole derivatives, highlighting their potential in therapeutic medicine for drug development. The structure-activity relationship (SAR) is also discussed to better understand their interactions and safety in biological systems. This work aims to expand on the reported chemistry and pharmacological potential of the thiadiazole moiety to validate their efficacy as promising pharmacophores in drug design and development.
Collapse
Affiliation(s)
- Benjamin Ayodipupo Babalola
- Department of Chemistry, Purdue University, 560 Oval Drive, IN 47907 West Lafayette, USA; Purdue Institute for Cancer Research, 201 S. University St., IN 47907 West Lafayette, USA.
| | - Lekhnath Sharma
- Department of Chemistry, Indian Institute of Technology Indore Simrol, Khandwa Road, Indore, India
| | - Olanike Olowokere
- Bioscience program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Monika Malik
- Department of Chemistry, Birla Institute of Technology & Science, Pilani, India
| | | |
Collapse
|
20
|
Mohammadi-Khanaposhtani M, Sayahi MH, Yazzaf R, Dastyafteh N, Halimi M, Iraji A, Dadgar A, Mojtabavi S, Faramarzi MA, Palimi M, Mirzazadeh R, Larijani B, Delnavazi MR, Mahdavi M. α-Glucosidase inhibition assay of galbanic acid and it amide derivatives: New excellent semi-synthetic α-glucosidase inhibitors. Bioorg Chem 2024; 150:107580. [PMID: 38959646 DOI: 10.1016/j.bioorg.2024.107580] [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: 02/22/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
α-Glucosidase inhibitory activity of galbanic acid and its new amide derivatives 3a-n were investigated. Galbanic acid and compounds 3a-n showed excellent anti-α-glucosidase activity with IC50 values ranging from 0.3 ± 0.3 μM to 416.0 ± 0.2 μM in comparison to positive control acarbose with IC50 value of = 750.0 ± 5.6. In the kinetic study, the most potent compound 3h demonstrated a competitive mode of inhibition with Ki = 0.57 µM. The interaction of the most potent compound 3h with the α-glucosidase was further elaborated by in vitro Circular dichroism assessment and in silico molecular docking and Molecular dynamics studies. Compound 3h was also non-cytotoxic on human normal cells. In silico study on pharmacokinetics and toxicity profile of the most potent galbanic acid derivatives demonstrated that these compounds are valuable lead compounds for further study in order to achieve new anti-diabetic agents.
Collapse
Affiliation(s)
- Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Hosein Sayahi
- Department of Chemistry, Payame Noor University, Tehran, Iran; Chemistry Department, College of Science, Shahid Chamran University of Ahvaz, Ahvaz 61357-4-3169, Iran
| | - Rozita Yazzaf
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Navid Dastyafteh
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Halimi
- Department of Biology, Babol Branch, Islamic Azad University, Babol, Iran
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Central Research Laboratory, Shiraz University of Medical Sciences, Shiraz, Iran; Liosa Pharmed Parseh Company, Shiraz, Iran
| | - Armin Dadgar
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdie Palimi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Delnavazi
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran; Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Chuang LH, Zhang H, Hong T, Xie S. Evaluating the Preferences and Willingness-to-Pay for Oral Antidiabetic Drugs Among Patients with Type 2 Diabetes Mellitus in China: A Discrete Choice Experiment. THE PATIENT 2024; 17:565-574. [PMID: 38642244 DOI: 10.1007/s40271-024-00694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/22/2024]
Abstract
PURPOSE To quantify the preferences for an oral antidiabetic drug (OAD) among patients with type 2 diabetes mellitus (T2DM) in China. METHODS A discrete choice experiment (DCE) with hypothetical OAD profiles was performed among patients with T2DM recruited from both online and offline sources. Each patient completed 12 DCE choice tasks. The attributes, elicited through mixed methods, include blood glucose level decrease, blood glucose level stability, frequency of medication, gastrointestinal side effects, dose adjustment and out-of-pocket expense. The conditional logit regression model was used to analyze the data. Patients' willingness-to-pay (WTP) was also calculated. Subgroup analyses based on patient characteristics were also conducted. RESULTS A total of 741 respondents were included in the analysis sample, covering 456 respondents online and 285 offline. The result showed that all attributes and levels were statistically significant, except one level "dose adjustment required for patients with hepatic or renal insufficiency" in the attribute of dose adjustment. WTP results showed that patients were willing to pay 12.06 and 23.20 yuan, respectively to reduce the frequency of medication from "once per day" and "three times per day" to "once every 2 weeks", respectively. Subgroup analyses showed that the frequency of medication (once versus two to three times per day) had the largest impact and influenced most coefficient estimates. CONCLUSION The results suggest that Chinese patients with T2DM prioritized better efficacy, less frequency of medication, lower gastrointestinal side effects, no dose adjustment required for patients with hepatic or renal insufficiency, and less out-of-pocket expense of OAD treatment.
Collapse
Affiliation(s)
- Ling-Hsiang Chuang
- Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden
- GongJing Healthcare (Nanjing) Co. Ltd, Nanjing, China
| | - Huanlan Zhang
- GongJing Healthcare (Nanjing) Co. Ltd, Nanjing, China
| | - Tianqi Hong
- School of Biomedical Engineering, McMaster University, Hamilton, Canada
| | - Shitong Xie
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
- Center for Social Science Survey and Data, Tianjin University, Tianjin, China.
| |
Collapse
|
22
|
Andavar M, Kamaraj R, Mahalingam Vijayakumar T, Murugesan A. Effectiveness of dual combination therapy of acarbose plus metformin and acarbose plus myo-inositol in ameliorating the metabolic and endocrinologic complications of polycystic ovary syndrome - A randomized controlled trial. Eur J Obstet Gynecol Reprod Biol 2024; 300:6-11. [PMID: 38972164 DOI: 10.1016/j.ejogrb.2024.07.001] [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: 03/04/2024] [Revised: 06/02/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION PCOS, beyond being characterized by reproductive disturbances, is a complicated rapid expanding metabolic and endocrinologic disorder of the recent times. Nearly 70% PCOS women show resistance to insulin. AIM The aim of the study is to determine and compare the effectiveness of acarbose plus metformin and acarbose plus myo-inositol combination therapy in alleviating the metabolic and endocrinologic complications of PCOS. MATERIALS AND METHODS An open labelled RCT was conducted on 168 PCOS women attending the gynaecology clinic at SRM MCH & RC, Chengalpattu and the trial was registered in CTRI (No. CTRI/2022/04/041877). Group A (n = 56) received metformin 500 mg/TID alone; group B (n = 54) received (acarbose 25 mg/TID for 4 weeks then 50 mg/TID for other 20 weeks) along with metformin 500 mg/TID and group C (n = 54) received (acarbose 25 mg/TID for 4 weeks then 50 mg/TID for other 20 weeks) along with myoinositol 1000 mg/BD. All parameters were measured at baseline and at the end of 6 months. RESULTS Significant reduction of LH, LH: FSH, TT, HOMA-IR was observed in all the groups. FSH increased only in metformin group. Increase in serum progesterone and reduction in FI, TGL, LDL were significant only in acarbose plus myo-inositol group. SHBG and HDL increased significantly only in acarbose plus metformin group. No changes in BMI, TC and VLDL were observed in any group. CONCLUSION Therefore, decrease in FI, HOMA-IR, TGL, LDL seen in acarbose plus myo-inositol group indirectly contributes to cardio-metabolic safety in PCOS. Similarly, a significant increase in SHBG levels with acarbose plus metformin group shows correction of the excess androgen and restoration of ovulation.
Collapse
Affiliation(s)
- Marina Andavar
- Department of Pharmacy Practice, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Raju Kamaraj
- Department of Pharmaceutical Regulatory Affairs, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India.
| | - Thangavel Mahalingam Vijayakumar
- Department of Pharmacy Practice, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Anuradha Murugesan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu 603 203, India
| |
Collapse
|
23
|
Mandal D, Sarmah JK, Harish V, Gupta J. Antioxidant, In Vitro Cytotoxicity, and Anti-diabetic Attributes of a Drug-Free Guar Gum Nanoformulation as a Novel Candidate for Diabetic Wound Healing. Mol Biotechnol 2024:10.1007/s12033-024-01261-z. [PMID: 39212825 DOI: 10.1007/s12033-024-01261-z] [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: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
The escalating intersection of diabetes and impaired wound healing poses a substantial societal burden, marked by an increasing prevalence of chronic wounds. Diabetic individuals struggle with hindered recovery, attributed to compromised blood circulation and diminished immune function, resulting in prolonged healing periods and elevated healthcare expenditures. To address this challenge, we report here a drug-free novel guar gum (GG)-based nano-formulation which is effective against diabetic wound healing. Nanoparticles with an average particle size of 32.4 nm display stability with negative zeta potential. Differential scanning calorimetry (DSC) and Fourier transform infrared (FTIR) analysis reveal alterations in thermal properties and molecular structures induced by the nano-particulation process. In vitro studies highlight the antioxidant potential of GGNP through concentration-dependent free radical scavenging activity in DPPH and ABTS assays. The nanoformulation also exhibits inhibitory effects on α-glucosidase and α-amylase enzymes. Cell viability studies have indicated moderate cytotoxicity in L929 cells and significant proliferation and migration in HaCaT cells, suggesting a positive impact on skin cells. In vitro enzymatic activity assessments under hyperglycaemic conditions reveal the potential of GGNP to modulate glutathione-S-transferase (GST), superoxide dismutase (SOD), and catalase activities as well as decreasing lipid peroxidation (LPO) levels, showcasing an antioxidant response. These results suggest GGNP as a promising candidate in diabetic wound healing.
Collapse
Affiliation(s)
- Debojyoti Mandal
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, Phagwara, Punjab, 144411, India
| | - Jayanta K Sarmah
- Department of Chemistry, Rabindranath Tagore University, Hojai, Assam, 782435, India.
| | - Vancha Harish
- School of Pharmaceutical Sciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, Phagwara, Punjab, 1444111, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, Phagwara, Punjab, 1444111, India.
| |
Collapse
|
24
|
Sepehri S, Farhadi G, Maghbul M, Nasiri F, Faramarzi MA, Mahnam K, Mojtabavi S, Mahdavi M, Moharrami Oranj Z. A new route to the synthesis of 2-hydrazolyl-4-thiazolidinone hybrids, evaluation of α-glucosidase inhibitory activity and molecular modeling insights. Heliyon 2024; 10:e36408. [PMID: 39247345 PMCID: PMC11380029 DOI: 10.1016/j.heliyon.2024.e36408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
One of the multifactorial worldwide health syndromes is diabetes mellitus which is increasing at a disturbing rate. The inhibition of α-glucosidase, an enzyme that catalyzes starch hydrolysis in the intestine, is one helpful therapeutic approach for controlling hyperglycemia related to type-2 diabetes. To discover α-glucosidase inhibitors, some 2-hydrazolyl-4-thiazolidinone hybrids (3a-e) were synthesized from new one-pot reaction procedures. Next, their chemical structures were confirmed by 1H NMR, 13C NMR, and FT-IR spectra, and elemental analysis technique. Then, the α-glucosidase inhibitory activity of the titled compounds was evaluated. Among them, derivatives 3b and 3c revealed the highest activity against α-glucosidase compared to acarbose as a drug. Enzyme kinetic studies of the most active derivative (3b) indicated a competitive inhibition. Finally, molecular modeling studies were accomplished to describe vital interactions of the most potent compounds (3b and 3c) with the α-glucosidase enzyme.
Collapse
Affiliation(s)
- Saghi Sepehri
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghazaleh Farhadi
- Students Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Maghbul
- Department of Applied Chemistry, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Farough Nasiri
- Department of Applied Chemistry, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Karim Mahnam
- Department of Biology, Faculty of Sciences, Shahrekord University, Shahrekord, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
25
|
Benjamin MAZ, Mohd Mokhtar RA, Iqbal M, Abdullah A, Azizah R, Sulistyorini L, Mahfudh N, Zakaria ZA. Medicinal plants of Southeast Asia with anti-α-glucosidase activity as potential source for type-2 diabetes mellitus treatment. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118239. [PMID: 38657877 DOI: 10.1016/j.jep.2024.118239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/13/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetes mellitus, a widespread chronic illness, affects millions worldwide, and its incidence is increasing alarmingly, especially in developing nations. Current pharmacological treatments can be costly and have undesirable side effects. To address this, medicinal plants with antidiabetic effects, particularly targeting α-glucosidase for controlling hyperglycaemia in type-2 diabetes mellitus (T2DM), hold promise for drug development with reduced toxicity and adverse reactions. AIM OF THIS REVIEW This review aims to succinctly collect information about medicinal plant extracts that exhibit antidiabetic potential through α-glucosidase inhibition using acarbose as a standard reference in Southeast Asia. The characteristics of this inhibition are based on in vitro studies. MATERIALS AND METHODS Relevant information on medicinal plants in Southeast Asia, along with α-glucosidase inhibition studies using acarbose as a positive control, was gathered from various scientific databases, including Scopus, PubMed, Web of Science, and Google Scholar. RESULTS About 49 papers were found from specific counties in Southeast Asia demonstrated notable α-glucosidase inhibitory potential of their medicinal plants, with several plant extracts showcasing activity comparable to or surpassing that of acarbose. Notably, 19 active constituents were identified for their α-glucosidase inhibitory effects. CONCLUSIONS The findings underscore the antidiabetic potential of the tested medicinal plant extracts, indicating their promise as alternative treatments for T2DM. This review can aid in the development of potent therapeutic medicines with increased effectiveness and safety for the treatment of T2DM.
Collapse
Affiliation(s)
- Mohammad Amil Zulhilmi Benjamin
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Ruzaidi Azli Mohd Mokhtar
- Biotechnology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Mohammad Iqbal
- Biotechnology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Azmahani Abdullah
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin, 21300, Kuala Nerus, Terengganu, Malaysia
| | - Roro Azizah
- Department of Environmental Health, Faculty of Public Health, Universitas Airlangga Kampus C, Jl. Dr. Ir. H. Soekarno, Mulyorejo, Kec. Mulyorejo, Kota Surabaya, Jawa Timur, 60115, Indonesia
| | - Lilis Sulistyorini
- Department of Environmental Health, Faculty of Public Health, Universitas Airlangga Kampus C, Jl. Dr. Ir. H. Soekarno, Mulyorejo, Kec. Mulyorejo, Kota Surabaya, Jawa Timur, 60115, Indonesia
| | - Nurkhasanah Mahfudh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Ahmad Dahlan, Jl. Prof. Dr. Soepomo Sh, Warungboto, Kec. Umbulharjo, Kota Yogyakarta, Daerah Istimewa Yogyakarta, 55164, Indonesia
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia; Department of Environmental Health, Faculty of Public Health, Universitas Airlangga Kampus C, Jl. Dr. Ir. H. Soekarno, Mulyorejo, Kec. Mulyorejo, Kota Surabaya, Jawa Timur, 60115, Indonesia; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Ahmad Dahlan, Jl. Prof. Dr. Soepomo Sh, Warungboto, Kec. Umbulharjo, Kota Yogyakarta, Daerah Istimewa Yogyakarta, 55164, Indonesia.
| |
Collapse
|
26
|
Lv SY, Cheng LP. Design, synthesis and inhibition evaluation of novel chalcone amide α-glucosidase inhibitors. Future Med Chem 2024; 16:1333-1345. [PMID: 39109435 PMCID: PMC11318676 DOI: 10.1080/17568919.2024.2347092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/09/2024] [Indexed: 08/15/2024] Open
Abstract
Aim: The purpose of this study is to design and synthesize a series of novel chalcone amide α-glucosidase (AG) inhibitors (L1-L10) based on virtual screening and molecular dynamics (MD) simulation. Materials & methods: Target compounds (L1-L10) were synthesized from 2-hydroxyacetophenone and methyl 4-formylbenzoate. Results: In vitro activity test shows that most compounds have good AG inhibition. Specially, compound L4 (IC50 = 8.28 ± 0.04 μM) had the best inhibitory activity, superior to positive control acarbose (IC50 = 8.36 ± 0.02 μM). Molecular docking results show that the good potency of L4 maybe attributed to strong interactions between chalcone skeleton and active site, and the torsion of carbon nitrogen bond in amide group. Conclusion: Compound L4 maybe regard as a good anti-Type II diabetes candidate to preform further study.
Collapse
Affiliation(s)
- Song Yao Lv
- School of Chemical & Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Li Ping Cheng
- School of Chemical & Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China
| |
Collapse
|
27
|
Ansariashlaghi S, Fakhrioliaei A, Mohammadi-Khanaposhtani M, Noori M, Asadi M, Mojtabavi S, Faramarzi MA, Esfahani EN, Rastegar H, Larijani B, Azizian H, Mahdavi M. New phenylthiosemicarbazide-phenoxy-1,2,3-triazole-N-phenylacetamides as dual inhibitors against α-glucosidase and PTP-1B for the treatment of type 2 diabetes. Arch Pharm (Weinheim) 2024; 357:e2300517. [PMID: 38593290 DOI: 10.1002/ardp.202300517] [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: 09/18/2023] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024]
Abstract
This study describes the design, synthesis, and evaluation of a novel series of phenylthiosemicarbazide-phenoxy-1,2,3-triazole-N-phenylacetamide derivatives (7a-l) as dual inhibitors of α-glucosidase and protein tyrosine phosphatase 1-B (PTB-1B). The latter enzymes are two important targets in the treatment of type 2 diabetes. The in vitro obtained data demonstrated that all title compounds 7a-l were more potent than the standard inhibitor acarbose against α-glucosidase while only four derivatives (7a, 7g, 7h, and 7h) were more potent than the standard inhibitor suramin against PTP-1B. Furthermore, these data showed that the most potent α-glucosidase inhibitor was compound 7i, with sixfold higher inhibitory activity than acarbose, and the most potent PTP-1B inhibitor was compound 7a with 3.5-fold higher inhibitory activity than suramin. Kinetic studies of compounds 7i and 7a revealed that they inhibited their target enzymes in a competitive mode. The docking study demonstrated that compounds 7i and 7a well occupied the active site pockets of α-glucosidase and PTP-1B, respectively. In silico pharmacokinetic and toxicity assays of the most potent compounds were performed, and the obtained results were compared with those of the standard inhibitors.
Collapse
Affiliation(s)
- Shirin Ansariashlaghi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Fakhrioliaei
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Milad Noori
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Asadi
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tabriz, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad A Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh N Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Rastegar
- Cosmetic Products Research Center, Iranian Food and Drug Administration, MOHE, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Andavar M, Kamaraj R, Vijayakumar TM, Murugesan A. Therapeutic potential of acarbose in ameliorating the metabolic and endocrinological complications of polycystic ovarian syndrome: a review. Curr Med Res Opin 2024; 40:1123-1135. [PMID: 38771729 DOI: 10.1080/03007995.2024.2358237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 05/17/2024] [Indexed: 05/23/2024]
Abstract
Polycystic ovarian syndrome is a perplexed condition addressing endocrinal, cardiometabolic and gynaecological issues. It affects women of adolescent age and is drastically increasing in the Indo-Asian ethnicity over the recent years. According to Rotterdam criteria, PCOS is characterized by clinical or biochemical excess androgen and polycystic ovarian morphology; however, it has been established in the recent years that PCOS exacerbates to further serious metabolic conditions on the long term. This is a narrative literature review and not systematic review and is based on PubMed searches with relevant keywords "Polycystic ovarian syndrome AND acarbose OR metformin OR myoinositol; PCOS AND metabolic syndrome OR cardiovascular disease OR menstrual irregularity OR infertility OR chronic anovulation OR clinical hyperandrogenism" used in the title and are limited to articles published in English language with no time limits. A prominent aspect of PCOS is hyperandrogenaemia and hyperinsulinemia. About 50-70% of afflicted women have compensatory hyperinsulinemia and close to one tierce suffer from anovulation and infertility. Insulin resistance leads to metabolic complications and works with luteinizing hormone in increasing the ovarian androgen production. This excess androgen leads to clinical manifestations, irregular menstrual cycles and infertility. There isn't an entire cure, only the symptomatic clinical factors are considered rather than focusing on the underlying long-term complications. Therefore, the article focuses on a potent alpha glucosidase inhibitor, acarbose which suppresses the post meal glucose and insulin by delaying the absorption of complex carbs. It exhibits cardio-metabolic and hormonal benefits and is well tolerable in the south asian population. This review highlights the safety, effectiveness of acarbose in ameliorating the long-term complications of PCOS.
Collapse
Affiliation(s)
- Marina Andavar
- SRM College of Pharmacy, Department of Pharmacy Practice, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Raju Kamaraj
- SRM College of Pharmacy, Department of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Thangavel Mahalingam Vijayakumar
- SRM College of Pharmacy, Department of Pharmacy Practice, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Anuradha Murugesan
- SRM Medical College Hospital and Research Centre, Department of Obstetrics and Gynaecology, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| |
Collapse
|
29
|
Zhang J, Lv J, Zhuang G, Zhang J, Hu F, Chen Y. The Evaluation of the Phytochemical Profiles and Antioxidant and α-Glucosidase Inhibitory Activities of Four Herbal Teas Originating from China: A Comparative Analysis of Aqueous and Ethanol Infusions. Foods 2024; 13:1705. [PMID: 38890931 PMCID: PMC11171806 DOI: 10.3390/foods13111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Recent research has demonstrated the positive impact of herbal tea consumption on postprandial blood glucose regulation. This study conducts a comparative analysis of aqueous and ethanol extractions on four herbal teas (Mallotus, Cyclocarya, Rubus, and Vine) to assess their phytochemical profiles and functional attributes. Phytochemical contents, antioxidant activities, α-glucosidase inhibitory activities, and chemical compositions are investigated via colorimetric analyses and UPLC-Q-Orbitrap HRMS/MS, respectively. Results indicate that Vine, among the teas studied, exhibits the most pronounced glucose-regulating effects under both extraction methods. While ethanol extractions yield higher phytochemical content overall, the compositions vary. Conversely, aqueous extracts demonstrate unexpectedly potent antioxidant activities and comparable α-glucosidase inhibitory activities to ethanol extracts. Phytochemical contents correlate positively with antioxidant activities and α-glucosidase inhibitory activities. However, antioxidant activities exhibit a weak positive correlation with α-glucosidase inhibitory activities. These findings provide evidence that aqueous extracts from herbal teas contain valuable phytochemical compositions beneficial for antioxidants and individuals with hyperglycemia, suggesting their potential as functional ingredients to enhance the nutritional value of herbal food products.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China (J.L.)
| | - Jinling Lv
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China (J.L.)
| | - Guodong Zhuang
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China (J.L.)
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao
| | - Junjia Zhang
- Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ 08901, USA
| | - Feng Hu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China (J.L.)
| | - Yongsheng Chen
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China (J.L.)
| |
Collapse
|
30
|
Khan H, Jan F, Shakoor A, Khan A, AlAsmari AF, Alasmari F, Ullah S, Al-Harrasi A, Khan M, Ali S. Design, synthesis, molecular docking study, and α-glucosidase inhibitory evaluation of novel hydrazide-hydrazone derivatives of 3,4-dihydroxyphenylacetic acid. Sci Rep 2024; 14:11410. [PMID: 38762658 PMCID: PMC11102520 DOI: 10.1038/s41598-024-62034-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: 12/24/2023] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
A series of novel Schiff base derivatives (1-28) of 3,4-dihydroxyphenylacetic acid were synthesized in a multi-step reaction. All the synthesized Schiff bases were obtained in high yields and their structures were determined by 1HNMR, 13CNMR, and HR-ESI-MS spectroscopy. Except for compounds 22, 26, 27, and 28, all derivatives show excellent to moderate α-glucosidase inhibition. Compounds 5 (IC50 = 12.84 ± 0.52 µM), 4 (IC50 = 13.64 ± 0.58 µM), 12 (IC50 = 15.73 ± 0.71 µM), 13 (IC50 = 16.62 ± 0.47 µM), 15 (IC50 = 17.40 ± 0.74 µM), 3 (IC50 = 18.45 ± 1.21 µM), 7 (IC50 = 19.68 ± 0.82 µM), and 2 (IC50 = 20.35 ± 1.27 µM) shows outstanding inhibition as compared to standard acarbose (IC50 = 873.34 ± 1.67 µM). Furthermore, a docking study was performed to find out the interaction between the enzyme and the most active compounds. With this research work, 3,4-dihydroxyphenylacetic acid Schiff base derivatives have been introduced as a potential class of α-glucosidase inhibitors that have remained elusive till now.
Collapse
Affiliation(s)
- Hammad Khan
- Organic Synthesis and Catalysis Research Laboratory, Institute of Chemical Sciences, University of Peshawar, Peshawar, 25120, Khyber Pakhtunkhwa, Pakistan
| | - Faheem Jan
- Shenyang National Laboratory for Materials Science, Institute of Metal Research Chinese Academy of Sciences, Shenyang, 110016, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Shenyang, 110016, Liaoning, China
| | - Abdul Shakoor
- Department of Chemistry, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, 616, Birkat Al Mauz, Nizwa, Oman
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, 616, Birkat Al Mauz, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, PO Box 33, 616, Birkat Al Mauz, Nizwa, Oman.
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| | - Shaukat Ali
- Organic Synthesis and Catalysis Research Laboratory, Institute of Chemical Sciences, University of Peshawar, Peshawar, 25120, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
31
|
Ahmed A, Zaib S, Bhat MA, Saeed A, Altaf MZ, Zahra FT, Shabir G, Rana N, Khan I. Acyl pyrazole sulfonamides as new antidiabetic agents: synthesis, glucosidase inhibition studies, and molecular docking analysis. Front Chem 2024; 12:1380523. [PMID: 38694406 PMCID: PMC11061460 DOI: 10.3389/fchem.2024.1380523] [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: 02/01/2024] [Accepted: 03/11/2024] [Indexed: 05/04/2024] Open
Abstract
Diabetes mellitus is a multi-systematic chronic metabolic disorder and life-threatening disease resulting from impaired glucose homeostasis. The inhibition of glucosidase, particularly α-glucosidase, could serve as an effective methodology in treating diabetes. Attributed to the catalytic function of glucosidase, the present research focuses on the synthesis of sulfonamide-based acyl pyrazoles (5a-k) followed by their in vitro and in silico screening against α-glucosidase. The envisaged structures of prepared compounds were confirmed through NMR and FTIR spectroscopy and mass spectrometry. All compounds were found to be more potent against α-glucosidase than the standard drug, acarbose (IC50 = 35.1 ± 0.14 µM), with IC50 values ranging from 1.13 to 28.27 µM. However, compound 5a displayed the highest anti-diabetic activity (IC50 = 1.13 ± 0.06 µM). Furthermore, in silico studies revealed the intermolecular interactions of most potent compounds (5a and 5b), with active site residues reflecting the importance of pyrazole and sulfonamide moieties. This interaction pattern clearly manifests various structure-activity relationships, while the docking results correspond to the IC50 values of tested compounds. Hence, recent investigation reveals the medicinal significance of sulfonamide-clubbed pyrazole derivatives as prospective therapeutic candidates for treating type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Atteeque Ahmed
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Zain Altaf
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Fatima Tuz Zahra
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ghulam Shabir
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Nehal Rana
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Imtiaz Khan
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
32
|
Hameed S, Saleem F, Özil M, Baltaş N, Salar U, Ashraf S, Ul-Haq Z, Taha M, Khan KM. Indenoquinoxaline-phenylacrylohydrazide hybrids as promising drug candidates for the treatment of type 2 diabetes: In vitro and in silico evaluation of enzyme inhibition and antioxidant activity. Int J Biol Macromol 2024; 263:129517. [PMID: 38266833 DOI: 10.1016/j.ijbiomac.2024.129517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/26/2023] [Accepted: 01/13/2024] [Indexed: 01/26/2024]
Abstract
Existing drugs that are being used to treat type-2 diabetes mellitus are associated with several side effects; thus, exploring potential drug candidates is still an utter need these days. Hybrids of indenoquinoxaline and hydrazide have never been explored as antidiabetic agents. In this study, a series of new indenoquinoxaline-phenylacrylohydrazide hybrids (1-30) were synthesized, structurally characterized, and evaluated for α-amylase and α-glucosidase inhibitory activities, as well as for their antioxidant properties. All scaffolds exhibited varying degrees of inhibitory activity against both enzymes, with IC50 values ranging from 2.34 to 61.12 μM for α-amylase and 0.42 to 54.72 μM for α-glucosidase. Particularly, compounds 10, 16, 17, 18, 24, and 25 demonstrated the highest efficacy in inhibiting α-amylase, while compounds 6, 7, 8, 10, 12, 14, 13, 16, 17, 18, 24, and 25 were the most effective α-glucosidase inhibitors, compared to standard acarbose. Moreover, most of these compounds displayed substantial antioxidant potential compared to standard butylated hydroxytoluene (BHT). Kinetics studies revealed competitive inhibition modes by compounds. Furthermore, a comprehensive in silico study and toxicity prediction were also conducted, further validating these analogs as potential drug candidates. The structured compounds demonstrated enhanced profiles, underscoring their potential as primary candidates in drug discovery.
Collapse
Affiliation(s)
- Shehryar Hameed
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Faiza Saleem
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Musa Özil
- Department of Chemistry, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Nimet Baltaş
- Department of Chemistry, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Uzma Salar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Pakistan Academy of Science, 3-Constitution Avenue, G-5/2, Islamabad 44000, Pakistan.
| |
Collapse
|
33
|
Wang H, Huang X, Pan Y, Zhang G, Tang S, Shao H, Jiao W. Synthesis and Biological Evaluation of New Dihydrofuro[3,2- b]piperidine Derivatives as Potent α-Glucosidase Inhibitors. Molecules 2024; 29:1179. [PMID: 38474691 DOI: 10.3390/molecules29051179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Inhibition of glycoside hydrolases has widespread application in the treatment of diabetes. Based on our previous findings, a series of dihydrofuro[3,2-b]piperidine derivatives was designed and synthesized from D- and L-arabinose. Compounds 32 (IC50 = 0.07 μM) and 28 (IC50 = 0.5 μM) showed significantly stronger inhibitory potency against α-glucosidase than positive control acarbose. The study of the structure-activity relationship of these compounds provides a new clue for the development of new α-glucosidase inhibitors.
Collapse
Affiliation(s)
- Haibo Wang
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhejiang Hongyuan Pharmaceutical Co., Ltd., Linhai 317016, China
| | - Xiaojiang Huang
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Pan
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoqing Zhang
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Senling Tang
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huawu Shao
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Wei Jiao
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| |
Collapse
|
34
|
Singh G, Singh R, Monga V, Mehan S. 3,5-Disubstituted-thiazolidine-2,4-dione hybrids as antidiabetic agents: Design, synthesis, in-vitro and In vivo evaluation. Eur J Med Chem 2024; 266:116139. [PMID: 38252989 DOI: 10.1016/j.ejmech.2024.116139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
Diabetes is one of the fastest-growing metabolic disorders, nearly doubling the number of patients each year. There are different treatment approaches available for the management of diabetes, which lacks due to their side effects. The inhibition of enzymes involved in the metabolism of complex polysaccharides to monosaccharides has proven beneficial in patients with type 2 diabetes mellitus. Two enzymes, α-amylase and α-glucosidase, have emerged as potential drug targets and are widely explored for drug development against type 2 diabetes mellitus. In this context, thiazolidine-2,4-diones (TZDs) have emerged as potential drug candidates for developing newer molecules against α-amylase and α-glucosidase. Nineteen TZD-hybrids were synthesized and evaluated in vitro α-amylase and α-glucosidase inhibitory activity. The compounds 7i, 7k, and 7p have emerged as the best dual inhibitors with IC50 of 10.33 ± 0.11-20.94 ± 0.76 μM and 10.19 ± 0.25-24.07 ± 1.56 μM against α-glucosidase and α-amylase, respectively. The derivatives had good anti-oxidant activity, displaying IC50 = 14.95 ± 0.65-23.27 ± 0.99 μM. The compounds 7k and 7p showed the best inhibition of reactive oxygen species in the PNAC-1 cells. The molecules exhibit good binding within the active site of α-amylase (PDB id: 1B2Y) and α-glucosidase (PDB id: 3W37), displaying binding energies of -7.5 to -10.7 kcal/mol and -7.4 to -10.3 kcal/mol, respectively. Further, the compounds were nontoxic (LD50 = 500-1311 mg/kg) and possessed good GI absorption. The compounds 7i, 7k, and 7p were evaluated in vivo antidiabetic activity in an STZ-induced diabetic model in Wistar rats. The compound 7p emerged as the best compound in the in vivo studies; however, the activity was lesser than that of the standard drug pioglitazone.
Collapse
Affiliation(s)
- Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, Punjab, India; Research Scholar, IK Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Rajveer Singh
- Department of Pharmacognosy, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, Punjab, India
| | - Vikramdeep Monga
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, Punjab, India.
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, Punjab, India, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, India).
| |
Collapse
|
35
|
Zhang T, Liu L, Chen Q, Wang Y, Gao X, Ma X, Yan P. Comparative Assessment of In Vitro Xanthine Oxidase and α-Glucosidase Inhibitory Activities of Cultured Cambial Meristematic Cells, Adventitious Roots, and Field-Cultivated Ginseng. Nutrients 2024; 16:443. [PMID: 38337727 PMCID: PMC10857066 DOI: 10.3390/nu16030443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Panax ginseng, a traditional Chinese medicine with a history spanning thousands of years, faces overexploitation and challenges related to extended growth periods. Tissue-cultured adventitious roots and stem cells are alternatives to wild and field-cultivated ginseng. In this study, we assessed the in vitro xanthine oxidase and α-glucosidase inhibitory activities of saponin extracts among cultured cambial meristematic cells (CMC), adventitious ginseng roots (AGR), and field-cultivated ginseng roots (CGR). The xanthine oxidase (XO) and α-glucosidase inhibitory activities were determined by uric acid estimation and the p-NPG method, respectively. Spectrophotometry and the Folin-Ciocalteu, aluminum nitrate, and Bradford methods were employed to ascertain the total saponins and phenolic, flavonoid, and protein contents. The calculated IC50 values for total saponin extracts against XO and α-glucosidase were 0.665, 0.844, and >1.6 mg/mL and 0.332, 0.745, and 0.042 mg/mL for AGR, CMC, CGR, respectively. Comparing the total saponin, crude protein, and total phenolic contents revealed that AGR > CMC > CGR. To the best of our knowledge, this study presents the first report on the in vitro comparison of xanthine oxidase and α-glucosidase inhibitory activities among AGR, CMC, and CGR. The findings offer valuable insights into the development of hypoglycemic and antihyperuricemic medicinal, nutraceutical, and functional products utilizing AGR and CMC.
Collapse
Affiliation(s)
- Tianhe Zhang
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China; (T.Z.); (Q.C.)
| | - Lijun Liu
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China; (T.Z.); (Q.C.)
| | - Qiqi Chen
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China; (T.Z.); (Q.C.)
| | - Yifei Wang
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China; (T.Z.); (Q.C.)
| | - Xiujun Gao
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China; (T.Z.); (Q.C.)
| | - Xingyi Ma
- School of Science, Harbin Institute of Technology, Shenzhen 518055, China
- Biosen International, Jinan 250117, China
- Briteley Institute of Life Sciences, Yantai 264003, China
- Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Peisheng Yan
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China; (T.Z.); (Q.C.)
| |
Collapse
|
36
|
Liu J, Li JH, Zhao SY, Chang YQ, Chen QX, Wu WF, Jiao SM, Xiao H, Zhang Q, Zhao JF, Xu J, Sun PH. Discovery of N-(phenylsulfonyl)thiazole-2-carboxamides as potent α-glucosidase inhibitors. Drug Dev Res 2024; 85:e22128. [PMID: 37984820 DOI: 10.1002/ddr.22128] [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: 08/06/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/22/2023]
Abstract
In a search for novel nonsugar α-glucosidase inhibitors for diabetes treatment, a series of N-(phenylsulfonyl)thiazole-2-carboxamide derivatives were designed and synthesized, the α-glucosidase inhibitory activities were then evaluated. Several compounds with promising α-glucosidase inhibitory effects were identified. Among these, compound W24 which shows low cytotoxicity and good α-glucosidase inhibitory activity with an IC50 value of 53.0 ± 7.7 μM, is more competitive compared with the commercially available drug acarbose (IC50 = 228.3 ± 9.2 μM). W24 was identified as a promising candidate in the development of α-glucosidase inhibitors. Molecular docking studies and molecular dynamics simulation were also performed to reveal the binding pattern of the active compound to α-glucosidase, and the binding free energy of the best compound W24 was 36.3403 ± 3.91 kcal/mol.
Collapse
Affiliation(s)
- Jun Liu
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Jia-Hao Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Si-Yu Zhao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Yi-Qun Chang
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Qiu-Xian Chen
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Wen-Fu Wu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Shu-Meng Jiao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Haichuan Xiao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Qiang Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Jian-Fu Zhao
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China
| | - Jun Xu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Ping-Hua Sun
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| |
Collapse
|
37
|
Kumar A, Mazumder R, Rani A, Pandey P, Khurana N. Novel Approaches for the Management of Type 2 Diabetes Mellitus: An Update. Curr Diabetes Rev 2024; 20:e051023221768. [PMID: 37888820 DOI: 10.2174/0115733998261903230921102620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 10/28/2023]
Abstract
Diabetes mellitus is an irreversible, chronic metabolic disorder indicated by hyperglycemia. It is now considered a worldwide pandemic. T2DM, a spectrum of diseases initially caused by tissue insulin resistance and slowly developing to a state characterized by absolute loss of secretory action of the β cells of the pancreas, is thought to be caused by reduced insulin secretion, resistance to tissue activities of insulin, or a combination of both. Insulin secretagogues, biguanides, insulin sensitizers, alpha-glucosidase inhibitors, incretin mimetics, amylin antagonists, and sodium-glucose co-transporter-2 (SGLT2) inhibitors are the main medications used to treat T2DM. Several of these medication's traditional dosage forms have some disadvantages, including frequent dosing, a brief half-life, and limited absorption. Hence, attempts have been made to develop new drug delivery systems for oral antidiabetics to ameliorate the difficulties associated with conventional dosage forms. In comparison to traditional treatments, this review examines the utilization of various innovative therapies (such as microparticles, nanoparticles, liposomes, niosomes, phytosomes, and transdermal drug delivery systems) to improve the distribution of various oral hypoglycemic medications. In this review, we have also discussed some new promising candidates that have been approved recently by the US Food and Drug Administration for the treatment of T2DM, like semaglutide, tirzepatide, and ertugliflozin. They are used as a single therapy and also as combination therapy with drugs like metformin and sitagliptin.
Collapse
Affiliation(s)
- Abhishek Kumar
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, UP 201306, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, UP 201306, India
| | - Anjna Rani
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, UP 201306, India
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP 201306, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
38
|
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
|
39
|
Liu BR, Shi XL, Yan JK, Zhao R. A high-resolution α-glucosidase inhibition profiling for targeted identification of natural antidiabetic products from Lycopodiella cernua (L.) Pic. Serm and their inhibitory mechanism study. Nat Prod Res 2023; 37:4099-4111. [PMID: 36710469 DOI: 10.1080/14786419.2023.2169860] [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: 08/01/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
The targeted identification of α-glucosidase inhibitors from the crude ethyl acetate of Lycopodiella cernua (L.) Pic. Serm (L.cernua) was guided by high-resolution inhibition profiling. The α-glucosidase inhibition profiling and HPLC-QTOF-MS showed tannins and serratenes were the corresponding antidiabetic constituents. Two new serratenes named 3β, 21β-dihydroxyserra-14-en-24-oic acid-3β-(4'-methoxy-5'-hydroxybenzoate) (4), 3β, 21α-dihydroxyserra-14-en-24-oic acid-3β-(4'-methoxy-5'-hydroxybenzoate) (7), together with two known compounds (5 and 6) were isolated. Their structures were elucidated by HR-ESI-MS and NMR. Compounds 5-7 inhibited the α-glucosidase activity in a non-competitive manner with Ki values ranging from 1.29 to 12.9 µM. The molecular docking result unveiled that 4-7 bound to the residues at the channel site, which enabled to block the substrate access. In addition, the molecular dynamics (MD) simulation of the most active compound 7 and α-glucosidase indicated the 4'-methoxy-5'-hydroxybenzoate group formed the stable hydrogen bonds and pi-pi T-shaped interactions with Arg312, Gln350 and Phe300 residues, while the rings D and E were stabilized by hydrophobic interaction.
Collapse
Affiliation(s)
- Bing-Rui Liu
- School of Public Heath, North China University of Science and Technology, Tangshan, China
| | - Xu-Liu Shi
- College of Chemistry and Technology, Hebei Agricultural University, Huanghua, China
| | - Jian-Kun Yan
- College of Chemistry and Technology, Hebei Agricultural University, Huanghua, China
| | - Rui Zhao
- Pharmacy Department, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Liu Y, Zhang J, An C, Liu C, Zhang Q, Ding H, Ma S, Xue W. Identification of Potential Mechanisms of Rk1 Combination with Rg5 in the Treatment of Type II Diabetes Mellitus by Integrating Network Pharmacology and Experimental Validation. Int J Mol Sci 2023; 24:14828. [PMID: 37834276 PMCID: PMC10573417 DOI: 10.3390/ijms241914828] [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: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
In this study, we aimed to explore the potential targets and functional mechanisms of Rk1 combined with Rg5 (Rk1+Rg5) against type II diabetes mellitus (T2DM). Network pharmacology and molecular docking were used to predict and verify the targets and signaling pathways of Rk1+Rg5 against T2DM. The results were further confirmed by a db/db mouse model and a model using PA-induced L6 cells. According to network pharmacology, a total of 250 core targets of Rk1+Rg5 towards T2DM were identified; the insulin resistance signaling pathways were enriched by KEGG. Results of molecular docking indicated good binding affinity of Rk1 and Rg5 to Akt1. In vivo and in vitro studies further showed that Rk1+Rg5 is an inhibitor of skeletal muscle insulin resistance. The results showed that Rk1+Rg5 significantly improved the hyperglycemic state of db/db mice, alleviated dyslipidemia, and promoted skeletal muscle glucose uptake. This phenomenon was closely related to the alleviation of the insulin resistance in skeletal muscles. Finally, the combination activated the Akt signaling pathway and promoted GLUT4 translocation to the cell membrane for glucose uptake. Altogether, our findings, for the first time, demonstrate that the combination of Rk1 and Rg5 could be beneficial for anti-T2DM, possibly involving ameliorated insulin resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wenjiao Xue
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Microbiology, Xiying Road 76, Xi’an 710043, China; (Y.L.); (J.Z.); (C.A.); (C.L.); (Q.Z.); (H.D.); (S.M.)
| |
Collapse
|
41
|
Kim JS, Kim HJ, Lee EB, Choi JH, Jung J, Jang HH, Park SY, Ha KC, Park YK, Joo JC, Lee SH. Supplementary Effects of Allium hookeri Extract on Glucose Tolerance in Prediabetic Subjects and C57BL/KsJ- db/db Mice. Pharmaceuticals (Basel) 2023; 16:1364. [PMID: 37895834 PMCID: PMC10610268 DOI: 10.3390/ph16101364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Allium hookeri (AH) has been used as a nutritional and medicinal food in Asia for many years. Our previous studies have described its anti-diabetic, anti-obesity, and anti-inflammatory activities in animal models and prediabetes. This study investigated whether AH could improve glycemia by modulating insulin secretion in prediabetic subjects through an in-depth study. Eighty prediabetic subjects (100 ≤ fasting plasma glucose < 140 mg/dL) were randomly assigned to a placebo (n = 40) group or an ethanol AH extract (500 mg/day, n = 40) group for 12 weeks. Dietary intake and physical activity, blood glucose (an oral glucose tolerance test for 120 min), insulin (insulin response to oral glucose for 120 min), area under the curve (AUC) of glucose or insulin after oral glucose intake, insulin sensitivity markers, C-peptide, adiponectin, glycated hemoglobin A1c (HbA1c) levels, hematological tests (WBC, RBC, hemoglobin, hematocrit, and platelet count), blood biochemical parameters (ALP, AST, total bilirubin, total protein, albumin, gamma-GT, BUN, creatinine, LD, CK, and hs-CRP), and urine parameters (specific gravity and pH) were examined at both baseline and 12 weeks after supplementation with placebo or AH capsules. Fifty-eight participants (placebo group: 20 men and 10 women; AH group: 13 men and 15 women) completed the study. AH supplementation moderately reduced postprandial blood glucose at 60 min (-6.14 mg/dL, p = 0.061), postprandial insulin levels at 90 min (-16.69 µU/mL, p = 0.017), the glucose AUC at 90 min (-412.52 mg*min/dL, p = 0.021), as well as the insulin AUC at 90 min (-978.77 µU*min/mL, p = 0.021) and 120 min (-1426.41 µU*min/mL, p = 0.015) when compared with the placebo group. However, there were no effects of AH on dietary intake and physical activity; HOMA index; HbAlc; C-peptide; or adiponectin, hematological-, blood biochemical-, and urinary markers. To confirm the effects of AH extract on blood glucose insulin sensitivity, C57BL/6J or C57BL/KsJ-db/db mice were used (n = 8/group). Body weight, fasting plasma glucose level, lipid profiles, liver and renal function, pancreatic histology, and insulin immunoreactivity were assessed. In the diabetic db/db mice, hyperglycemia, which was accompanied by an increase in insulin secretion in diabetic mice, was significantly reduced by AH treatment, resulting in the alleviation of β-cell overcompensation and insulin resistance. We confirmed that AH supplementation can effectively control blood glucose and insulin levels by improving insulin sensitivity and may be a potential agent for glycemic control in subjects with prediabetes and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ji-Su Kim
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| | - Hyun-Ju Kim
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju 61755, Jeolla, Republic of Korea;
| | - Eun-Byeol Lee
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| | - Ji-Hye Choi
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| | - Jieun Jung
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| | - Hwan-Hee Jang
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| | - Shin-Young Park
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| | - Ki-Chan Ha
- Healthcare Claims & Management Incorporation, Jeonju 54858, Jeonbuk, Republic of Korea; (K.-C.H.); (Y.-K.P.)
| | - Yu-Kyung Park
- Healthcare Claims & Management Incorporation, Jeonju 54858, Jeonbuk, Republic of Korea; (K.-C.H.); (Y.-K.P.)
| | - Jong-Cheon Joo
- Department of Sasang Constitutional Medicine, College of Korean Medicine, Wonkwang University, Iksan 54596, Jeonbuk, Republic of Korea;
| | - Sung-Hyen Lee
- Functional Food Division, Department of Agro-food Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Jeonbuk, Republic of Korea; (J.-S.K.); (E.-B.L.); (J.-H.C.); (J.J.); (H.-H.J.); (S.-Y.P.)
| |
Collapse
|
42
|
Zhu D, Yuan Z, Wu D, Wu C, El-Seedi HR, Du M. The dual-function of bioactive peptides derived from oyster (Crassostrea gigas) proteins hydrolysates. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
43
|
Wang L, Ai C, Jin C, Mou J, Deng Y. Xanthones as potential α-glucosidase non-competition inhibitors: Synthesis, inhibitory activities, and in silico studies. Chem Biol Drug Des 2023; 102:547-556. [PMID: 37249098 DOI: 10.1111/cbdd.14272] [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: 02/23/2023] [Revised: 04/12/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
α-glucosidase inhibitors (AGIs) were commonly used in clinical for the treatment of type 2 diabetes. Xanthones were naturally occurring antioxidants, and they may also be potential AGIs. In this study, eleven 1,6- and 1,3-substituted xanthone compounds were designed and synthesized, of which four were new compounds. Their α-glucosidase inhibitory activities in vitro and in silico were evaluated. Five xanthone compounds with higher activity than acarbose were screened out, and the xanthones substituted at the 1,6-positions were more likely to be potential α-glucosidase non-competitive inhibitors. The binding mode of xanthones with α-glucosidase was further studied by molecular docking method, and the results showed that the inhibitory effect of non-competitive inhibitors on site 1 of α-glucosidase may be related to the hydrogen bonds formed by the compounds with amino acid residues ASN165, HIS209, TRY207, ASP243, and SER104. This study provided a theoretical basis of the rapid discovery and structural modification of non-competitive xanthone inhibitors of α-glucosidase.
Collapse
Affiliation(s)
- Lili Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China
| | - Chenyang Ai
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Chaoqun Jin
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Jiajia Mou
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yanru Deng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
44
|
Álvarez-Almazán S, Solís-Domínguez LC, Duperou-Luna P, Fuerte-Gómez T, González-Andrade M, Aranda-Barradas ME, Palacios-Espinosa JF, Pérez-Villanueva J, Matadamas-Martínez F, Miranda-Castro SP, Mercado-Márquez C, Cortés-Benítez F. Anti-Diabetic Activity of Glycyrrhetinic Acid Derivatives FC-114 and FC-122: Scale-Up, In Silico, In Vitro, and In Vivo Studies. Int J Mol Sci 2023; 24:12812. [PMID: 37628991 PMCID: PMC10454726 DOI: 10.3390/ijms241612812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Type 2 diabetes (T2D) is one of the most common diseases and the 8th leading cause of death worldwide. Individuals with T2D are at risk for several health complications that reduce their life expectancy and quality of life. Although several drugs for treating T2D are currently available, many of them have reported side effects ranging from mild to severe. In this work, we present the synthesis in a gram-scale as well as the in silico and in vitro activity of two semisynthetic glycyrrhetinic acid (GA) derivatives (namely FC-114 and FC-122) against Protein Tyrosine Phosphatase 1B (PTP1B) and α-glucosidase enzymes. Furthermore, the in vitro cytotoxicity assay on Human Foreskin fibroblast and the in vivo acute oral toxicity was also conducted. The anti-diabetic activity was determined in streptozotocin-induced diabetic rats after oral administration with FC-114 or FC-122. Results showed that both GA derivatives have potent PTP1B inhibitory activity being FC-122, a dual PTP1B/α-glucosidase inhibitor that could increase insulin sensitivity and reduce intestinal glucose absorption. Molecular docking, molecular dynamics, and enzymatic kinetics studies revealed the inhibition mechanism of FC-122 against α-glucosidase. Both GA derivatives were safe and showed better anti-diabetic activity in vivo than the reference drug acarbose. Moreover, FC-114 improves insulin levels while decreasing LDL and total cholesterol levels without decreasing HDL cholesterol.
Collapse
Affiliation(s)
- Samuel Álvarez-Almazán
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Luz Cassandra Solís-Domínguez
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Paulina Duperou-Luna
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Teresa Fuerte-Gómez
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Martin González-Andrade
- Laboratory of Biosensors and Molecular Modelling, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - María E. Aranda-Barradas
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Juan Francisco Palacios-Espinosa
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Jaime Pérez-Villanueva
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Félix Matadamas-Martínez
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Susana Patricia Miranda-Castro
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Crisóforo Mercado-Márquez
- Isolation and Animal Facility Unit, Facultad de Estudios Superiores Cuautitlán 28, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54714, Mexico;
| | - Francisco Cortés-Benítez
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| |
Collapse
|
45
|
Auti PS, Jagetiya S, Paul AT. Chromone Containing Hybrid Analogs: Synthesis and Applications in Medicinal Chemistry. Chem Biodivers 2023; 20:e202300587. [PMID: 37332056 DOI: 10.1002/cbdv.202300587] [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: 04/24/2023] [Revised: 06/17/2023] [Accepted: 06/17/2023] [Indexed: 06/20/2023]
Abstract
The use of privileged scaffolds has proven beneficial for generating novel bioactive scaffolds in drug discovery program. Chromone is one such privileged scaffold that has been exploited for designing pharmacologically active analogs. The molecular hybridization technique combines the pharmacophoric features of two or more bioactive compounds to avail a better pharmacological activity in the resultant hybrid analogs. The current review summarizes the rationale and techniques involved in developing hybrid analogs of chromone, which show potential in fields of obesity, diabetes, cancer, Alzheimer's disease and microbial infections. Here the molecular hybrids of chromone with various pharmacologically active analogs or fragments (donepezil, tacrine, pyrimidines, azoles, furanchalcones, hydrazones, quinolines, etc.) are discussed with their structure-activity relationship against above-mentioned diseases. Detailed methodologies for the synthesis of corresponding hybrid analogs have also been described, with suitable synthetic schemes. The current review will shed light on various strategies utilized for the design of hybrid analogs in the field of drug discovery. The importance of hybrid analogs in various disease conditions is also illustrated.
Collapse
Affiliation(s)
- Prashant S Auti
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sakshi Jagetiya
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Atish T Paul
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| |
Collapse
|
46
|
Fujiwara A, Kanda S, Ueno K, Fujie H, Sekine N. Reactive hypoglycemia owing to an intrahepatic congenital portosystemic shunt in an older patient. Diabetol Int 2023; 14:298-303. [PMID: 37397900 PMCID: PMC10307763 DOI: 10.1007/s13340-023-00627-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/02/2023] [Indexed: 07/04/2023]
Abstract
An 85-year-old woman was admitted to our hospital because of hypoglycemia and impairment of consciousness several hours after breakfast. Because the hypoglycemia predominantly occurred 2-4 h after meals, we diagnosed reactive hypoglycemia. An oral glucose tolerance test showed prolonged hyperinsulinemia following the postprandial hyperglycemia, with a subsequent rapid decrease in blood glucose concentration. The post-stimulus plasma C-peptide concentration was relatively low compared to the plasma insulin concentration. Abdominal computed tomography revealed an intrahepatic congenital portosystemic shunt (CPSS). On the basis of these findings, we concluded that the reactive hypoglycemia was induced by the CPSS, via a reduction in hepatic insulin extraction. Treatment with an alpha-glucosidase inhibitor resolved the reactive hypoglycemia. CPSS comprises anomalous vascular connections between the portal vein and the systemic venous circulation, and reactive hypoglycemia is a rare complication of this malformation, which has most frequently been reported in children, with only a few cases reported in adults. However, this case indicates that even in adult patients, imaging studies should be conducted to rule out CPSS as the cause of the reactive hyperglycemia.
Collapse
Affiliation(s)
- Akiko Fujiwara
- Department of Diabetology and Endocrinology, JCHO Tokyo Shinjuku Medical Center, 5-1, Tsukudo-cho, Shinjuku-ku, Tokyo, 162-8543 Japan
- Department of Diabetes and Metabolic Diseases, The University of Tokyo Hospital, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Shuhei Kanda
- Department of Diabetology and Endocrinology, JCHO Tokyo Shinjuku Medical Center, 5-1, Tsukudo-cho, Shinjuku-ku, Tokyo, 162-8543 Japan
| | - Keisuke Ueno
- Department of Diabetology and Endocrinology, JCHO Tokyo Shinjuku Medical Center, 5-1, Tsukudo-cho, Shinjuku-ku, Tokyo, 162-8543 Japan
| | - Hajime Fujie
- Department of Gastroenterology, JCHO Tokyo Shinjuku Medical Center, 5-1, Tsukudo-cho, Shinjuku-ku, Tokyo, 162-8543 Japan
| | - Nobuo Sekine
- Department of Diabetology and Endocrinology, JCHO Tokyo Shinjuku Medical Center, 5-1, Tsukudo-cho, Shinjuku-ku, Tokyo, 162-8543 Japan
| |
Collapse
|
47
|
Boers HM, van Dijk TH, Duchateau GS, Mela DJ, Hiemstra H, Hoogenraad AR, Priebe MG. Effect of mulberry fruit extract on glucose fluxes after a wheat porridge meal: a dual isotope study in healthy human subjects. Eur J Clin Nutr 2023; 77:741-747. [PMID: 36944719 DOI: 10.1038/s41430-023-01282-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/04/2023] [Accepted: 03/09/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Previous research has shown the efficacy of mulberry extracts for lowering post-prandial glucose (PPG) responses. The postulated mechanism is slowing of glucose absorption, but effects on glucose disposal or endogenous production are also possible. This research assessed the effect of a specified mulberry fruit extract (MFE) on these three glucose flux parameters. METHODS The study used a double-blind, randomized, controlled, full cross-over design. In 3 counter-balanced treatments, 12 healthy adult male subjects, mean (SD) age 24.9 (2.50) years and body mass index 22.5 (1.57) kg/m2, consumed porridge prepared from 13C-labelled wheat, with or without addition of 0.75 g MFE, or a solution of 13C-glucose in water. A co-administered 2H-glucose venous infusion allowed for assessment of glucose disposal. Glucose flux parameters, cumulative absorption (time to 50% absorption, T50%abs), and PPG positive incremental area under the curve from 0 to 120 min (+iAUC0-120) were determined from total and isotopically labelled glucose in plasma. As this exploratory study was not powered for formal inferential statistical tests, results are reported as the mean percent difference (or minutes for T50%abs) between treatments with 95% CI. RESULTS MFE increased mean T50%abs by 10.2 min, (95% CI 3.9-16.5 min), and reduced mean 2 h post-meal rate of glucose appearance by 8.4% (95% CI -14.9 to -1.4%) and PPG + iAUC0-120 by 11% (95% CI -26.3 to -7.3%), with no significant changes in glucose disposal or endogenous production. CONCLUSIONS The PPG-lowering effect of MFE is primarily mediated by a reduced rate of glucose uptake.
Collapse
Affiliation(s)
- Hanny M Boers
- Unilever Foods Innovation Centre, 6708 WH, Wageningen, The Netherlands.
| | - Theo H van Dijk
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Guus S Duchateau
- Unilever Foods Innovation Centre, 6708 WH, Wageningen, The Netherlands
| | - David J Mela
- Unilever Foods Innovation Centre, 6708 WH, Wageningen, The Netherlands.
| | - Harry Hiemstra
- Unilever Foods Innovation Centre, 6708 WH, Wageningen, The Netherlands
| | | | - Marion G Priebe
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
48
|
Nguyen NH, Duong TH, Truong Nguyen H, Vu YT, Tran TMD, Ho TTV, Mai CC, Mai DT, Nguyen HC, Thuy Le H, Pham DD. New Halogenated Flavonoids from Adenosma bracteosum and Vitex negundo and Their α-Glucosidase Inhibition. Chem Biodivers 2023; 20:e202300390. [PMID: 37293748 DOI: 10.1002/cbdv.202300390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
Adenosma bracteosum and Vitex negundo are natural sources of methoxylated flavonoids. Little is known about the α-glucosidase inhibition of multi-methoxylated flavonoid derivatives. Eighteen natural flavonoids were isolated from A. bracteosum and V. negundo. Seven halogenated derivatives were synthesized. Their chemical structures were elucidated by extensive NMR analysis and high-resolution mass spectroscopy as well as comparisons in literature. All compounds were evaluated for their α-glucosidase inhibition. Most compounds showed good activity with IC50 values ranging from 16.7 to 421.8 μM. 6,8-Dibromocatechin was the most active compound with an IC50 value of 16.7 μM. A molecular docking study was conducted, indicating that those compounds are potent α-glucosidase inhibitors.
Collapse
Affiliation(s)
- Ngoc-Hong Nguyen
- CirTech Institute, HUTECH University, 475 A Dien Bien Phu Street, Binh Thanh District, Ho Chi Minh City, 72300, Vietnam
| | - Thuc-Huy Duong
- Department of Chemistry, Ho Chi Minh City University of Education, 280 An Duong Vuong Street, District 5, Ho Chi Minh City, 72700, Vietnam
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 72900, Vietnam
| | - Y Thien Vu
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 72900, Vietnam
| | - Thi-Minh-Dinh Tran
- Department of Biology, Ho Chi Minh City University of Education, 280 An Duong Vuong Street, District 5, Ho Chi Minh City, 72700, Vietnam
| | - Thi-Thanh-Van Ho
- Department of Chemistry, Ho Chi Minh City University of Education, 280 An Duong Vuong Street, District 5, Ho Chi Minh City, 72700, Vietnam
| | - Chi-Cong Mai
- Department of Chemistry, Ho Chi Minh City University of Education, 280 An Duong Vuong Street, District 5, Ho Chi Minh City, 72700, Vietnam
| | - Dinh-Tri Mai
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, 122000, Vietnam
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, 1A TL29 Street, Thanh Loc ward, District 12, Ho Chi Minh City, Vietnam
| | - Hoang-Chuong Nguyen
- Faculty of Biology-Biotechnology, University of Science, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, 72711, Vietnam
| | - Huong Thuy Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 72900, Vietnam
| | - Duc-Dung Pham
- Department of Chemistry, Ho Chi Minh City University of Education, 280 An Duong Vuong Street, District 5, Ho Chi Minh City, 72700, Vietnam
| |
Collapse
|
49
|
Erukainure OL, Otukile KP, Harejane KR, Salau VF, Aljoundi A, Chukwuma CI, Matsabisa MG. Computational insights into the antioxidant and antidiabetic mechanisms of cannabidiol: An in vitro and in silico study. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
|
50
|
Ali M, Malik K, Zaidi A, Farooq U, Bukhari SM, Majeed Z, Mahnashi MH, Nawazish S, Abdulwahab A, Alshaibari KS. In-vitro high-throughput library screening-Kinetics and molecular docking studies of potent inhibitors of α-glucosidase. PLoS One 2023; 18:e0286159. [PMID: 37390110 PMCID: PMC10313066 DOI: 10.1371/journal.pone.0286159] [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: 01/30/2023] [Accepted: 05/09/2023] [Indexed: 07/02/2023] Open
Abstract
High throughput screening of synthetic compounds against vital enzymes is the way forward for the determination of potent enzyme inhibitors. In-vitro high throughput library screening of 258 synthetic compounds (comp. 1-258), was performed against α-glucosidase. The active compounds out of this library were investigated for their mode of inhibition and binding affinities towards α-glucosidase through kinetics as well as molecular docking studies. Out of all the compounds selected for this study, 63 compounds were found active within the IC50 range of 3.2 μM to 50.0 μM. The most potent inhibitor of α-glucosidase out of this library was the derivative of an oxadiazole (comp. 25). It showed the IC50 value of 3.23 ± 0.8 μM. Other highly active compounds were the derivatives of ethyl-thio benzimidazolyl acetohydrazide with IC50 values of 6.1 ± 0.5 μM (comp. 228), 6.84 ± 1.3 μM (comp. 212), 7.34 ± 0.3 μM (comp. 230) and 8.93 ± 1.0 μM (comp. 210). For comparison, the standard (acarbose) showed IC50 = 378.2 ± 0.12 μM. Kinetic studies of oxadiazole (comp. 25) and ethylthio benzimidazolyl acetohydrazide (comp. 228) derivatives indicated that Vmax and Km, both change with changing concentrations of inhibitors which suggests an un-competitive mode of inhibition. Molecular docking studies of these derivatives with the active site of α-glucosidase (PDB ID:1XSK), revealed that these compounds mostly interact with acidic or basic amino acid residues through conventional hydrogen bonds along with other hydrophobic interactions. The binding energy values of compounds 25, 228, and 212 were -5.6, -8.7 and -5.4 kcal.mol-1 whereas RMSD values were 0.6, 2.0, and 1.7 Å, respectively. For comparison, the co-crystallized ligand showed a binding energy value of -6.6 kcal.mol-1 along with an RMSD value of 1.1 Å. Our study predicted several series of compounds as active inhibitors of α-glucosidase including some highly potent inhibitors.
Collapse
Affiliation(s)
- Majid Ali
- Department of Chemistry, COMSATS University Islamabad, KPK, Abbottabad, Pakistan
- Department of Chemistry, Higher Education Department, Government Postgraduate College No.1, Abbottabad, KP, Pakistan
| | - Khuram Malik
- Department of Chemistry, COMSATS University Islamabad, KPK, Abbottabad, Pakistan
| | - Asma Zaidi
- Department of Chemistry, COMSATS University Islamabad, KPK, Abbottabad, Pakistan
| | - Umar Farooq
- Department of Chemistry, COMSATS University Islamabad, KPK, Abbottabad, Pakistan
| | - Syed Majid Bukhari
- Department of Chemistry, COMSATS University Islamabad, KPK, Abbottabad, Pakistan
| | - Zahid Majeed
- Faculty of Science, Department of Biotechnology, The University of Azad Jammu and Kashmir, Chehla Campus, Muzaffarabad, Pakistan
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Shamyla Nawazish
- Department of Environmental Sciences, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | | | | |
Collapse
|