ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES
ADDITION / CORRECTIONThis article has been corrected. View the notice.

α-Glucosidase Inhibitors from Brickellia cavanillesii

View Author Information
Facultad de Química, Universidad Nacional Autónoma de México, México DF 04510, México
Instituto Nacional de Medicina Genómica, Secretaría de Salud, México DF 14610, México
§ Instituto de Biología, Universidad Nacional Autónoma de México, México DF 04510, México
*Tel: (525)-55-622-5289. Fax: (525)-55-622-5329. E-mail: [email protected]
Cite this: J. Nat. Prod. 2012, 75, 5, 968–974
Publication Date (Web):May 15, 2012
https://doi.org/10.1021/np300204p
Copyright © 2012 The American Chemical Society and American Society of Pharmacognosy

    Article Views

    3311

    Altmetric

    -

    Citations

    LEARN ABOUT THESE METRICS
    Other access options
    Supporting Info (1)»

    Abstract

    Abstract Image

    An aqueous extract from the aerial parts of Brickellia cavanillesii attenuated postprandial hyperglycemia in diabetic mice during oral glucose and sucrose tolerance tests. Experimental type-II DM was achieved by treating mice with streptozotocin (100 mg/kg) and β-nicotinamide adenine dinucleotide (40 mg/kg). These pharmacological results demonstrated that B. cavanillesii is effective for controlling fasting and postprandial blood glucose levels in animal models. The same aqueous extract also showed potent inhibitory activity (IC50 = 0.169 vs 1.12 mg/mL for acarbose) against yeast α-glucosidase. Bioassay-guided fractionation of the active extract using the α-glucosidase inhibitory assay led to the isolation of several compounds including two chromenes [6-acetyl-5-hydroxy-2,2-dimethyl-2H-chromene (1) and 6-hydroxyacetyl-5-hydroxy-2,2-dimethyl-2H-chromene (2)], two sesquiterpene lactones [caleins B (3) and C (4)], several flavonoids [acacetin (5), genkwanin (6), isorhamnetin (7), kaempferol (8), and quercetin (9)], and 3,5-di-O-caffeoylquinic acid (10). Chromene 2 is a new chemical entity. Compounds 2, 4, 7, and 9 inhibited the activity of yeast α-glucosidase with IC50 0.42, 0.28, 0.16, and 0.53 mM, respectively, vs 1.7 mM for acarbose. Kinetic analysis revealed that compounds 4 and 7 behaved as mixed-type inhibitors with Ki values of 1.91 and 0.41 mM, respectively, while 2 was noncompetititive, with a Ki of 0.13 mM. Docking analysis predicted that these compounds, except 2, bind to the enzyme at the catalytic site.

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. You can change your affiliated institution below.

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    Effect of an infusion of B. cavanillesii on blood glucose levels of normal and STZ-diabetic mice. Effect of an infusion of B. cavanillesii on blood glucose levels of normal and STZ-diabetic mice after an oral load of glucose (1.5 g/kg). Effect of an infusion of B. cavanillesii on blood glucose levels of normal and STZ-diabetic mice after a load of sucrose (3 g/kg). Effect of an infusion of B. cavanillesii on blood glucose levels in NAD-STZ-diabetic mice after an oral load of glucose (1.5 g/kg). Effect of an infusion of B. cavanillesii on blood glucose levels in NAD-STZ-diabetic mice after an oral load of sucrose (3 g/kg). 13C and 1HNMR, DEPT, NOESY, HMBC, and HSQC spectra for compound 2. Lineweaver–Burk and Dixon plots of α-glucosidase inhibition at different concentrations of substrate and inhibitors for acarbose. Lineweaver–Burk and Dixon plots of α-glucosidase inhibition at different concentrations of substrate and inhibitors for 7. This material is available free of charge via the Internet at http://pubs.acs.org.

    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

    Cited By

    This article is cited by 91 publications.

    1. Soheila Esmaili, Ahmad Ebadi, Ardeshir Khazaei, Hamideh Ghorbani, Mohammad Ali Faramarzi, Somayeh Mojtabavi, Mohammad Mahdavi, Zahra Najafi. Novel Pyrano[3,2-c]quinoline-1,2,3-triazole Hybrids as Potential Anti-Diabetic Agents: In Vitro α-Glucosidase Inhibition, Kinetic, and Molecular Dynamics Simulation. ACS Omega 2023, 8 (26) , 23412-23424. https://doi.org/10.1021/acsomega.3c00133
    2. Norika Miyazaki, Kazutaka Sanada, Takumi Nakamura, Aoi Washio, Yasushi Yoshida, Takashi Mino, Yoshio Kasashima, Masami Sakamoto. Behavior of All Chiral Standard Amino Acids for Chiral Symmetry Breaking of p-Anisoin. Crystal Growth & Design 2022, 22 (8) , 4673-4679. https://doi.org/10.1021/acs.cgd.2c00480
    3. Chloe N. Larcombe, Lara R. Malins. Accessing Diverse Cross-Benzoin and α-Siloxy Ketone Products via Acyl Substitution Chemistry. The Journal of Organic Chemistry 2022, 87 (14) , 9408-9413. https://doi.org/10.1021/acs.joc.2c00801
    4. Jiuzhong Huang, Jianxiao Li, Jia Zheng, Wanqing Wu, Weigao Hu, Lu Ouyang, and Huanfeng Jiang . Dual Role of H2O2 in Palladium-Catalyzed Dioxygenation of Terminal Alkenes. Organic Letters 2017, 19 (13) , 3354-3357. https://doi.org/10.1021/acs.orglett.7b01228
    5. Yan-Yan Ma, Deng-Gao Zhao, Ai-Yu Zhou, Yu Zhang, Zhiyun Du, and Kun Zhang . α-Glucosidase Inhibition and Antihyperglycemic Activity of Phenolics from the Flowers of Edgeworthia gardneri. Journal of Agricultural and Food Chemistry 2015, 63 (37) , 8162-8169. https://doi.org/10.1021/acs.jafc.5b03081
    6. Araceli Pérez-Vásquez, Erika Castillejos-Ramírez, Sol Cristians, and Rachel Mata . Development of a UHPLC-PDA Method for the Simultaneous Quantification of 4-Phenylcoumarins and Chlorogenic Acid in Exostema caribaeum Stem Bark. Journal of Natural Products 2014, 77 (3) , 516-520. https://doi.org/10.1021/np400785z
    7. Rachel Mata, Sol Cristians, Sonia Escandón-Rivera, Krutzkaya Juárez-Reyes, and Isabel Rivero-Cruz . Mexican Antidiabetic Herbs: Valuable Sources of Inhibitors of α-Glucosidases. Journal of Natural Products 2013, 76 (3) , 468-483. https://doi.org/10.1021/np300869g
    8. Thua-Phong Lam, Ngoc-Vi Nguyen Tran, Long-Hung Dinh Pham, Nghia Vo-Trong Lai, Bao-Tran Ngoc Dang, Ngoc-Lam Nguyen Truong, Song-Ky Nguyen-Vo, Thuy-Linh Hoang, Tan Thanh Mai, Thanh-Dao Tran. Flavonoids as dual-target inhibitors against α-glucosidase and α-amylase: a systematic review of in vitro studies. Natural Products and Bioprospecting 2024, 14 (1) https://doi.org/10.1007/s13659-023-00424-w
    9. Saurabh Singh, Subhasish Ray, Gaurav Shukla, Malkeet Singh, Maya Shankar Singh. Electrochemical Hydroxylation of α‐ Bromoacetophenones: Access to α‐ Hydroxyacetophenones. Advanced Synthesis & Catalysis 2024, https://doi.org/10.1002/adsc.202301395
    10. Shivam, Asheesh Kumar Gupta, Sushil Kumar. Review on Diabetic Complications and their Management by Flavonoids and Triterpenoids. The Natural Products Journal 2023, 13 (8) https://doi.org/10.2174/2210315513666230330082412
    11. Qinhao Guan, Lihua Tang, Liangliang Zhang, Lixin Huang, Man Xu, Yuan Wang, Meng Zhang. Molecular insights into α‐glucosidase inhibition and antiglycation properties affected by the galloyl moiety in (−)‐epigallocatechin‐3‐gallate. Journal of the Science of Food and Agriculture 2023, 103 (15) , 7381-7392. https://doi.org/10.1002/jsfa.12818
    12. Naoual El Menyiy, Sara Aboulaghras, Saad Bakrim, Rania Moubachir, Doaue Taha, Asaad Khalid, Ashraf N. Abdalla, Alanood S. Algarni, Andi Hermansyah, Long Chiau Ming, Marius Emil Rusu, Abdelhakim Bouyahya. Genkwanin: An emerging natural compound with multifaceted pharmacological effects. Biomedicine & Pharmacotherapy 2023, 165 , 115159. https://doi.org/10.1016/j.biopha.2023.115159
    13. Jinfang Xu, Zhiyu Fan, Gangqiang Yang, Yanan Yang, Xinjie Wu, Tongtong Li, Qili Wang, Jianping Gao. Identification of yeast α -glucosidase inhibitors from Pueraria lobata by ligand fishing based on magnetic mesoporous silicon combined with knock-out/knock-in technology. Food & Function 2023, 14 (4) , 1952-1961. https://doi.org/10.1039/D2FO03475A
    14. Bui Thanh Tung, Nguyen Bao Kim, Nguyen Nhu Son. Natural Compounds for Diabetes Prevention and Treatment. 2023, 131-146. https://doi.org/10.4018/978-1-6684-6737-4.ch008
    15. Kunle Okaiyeto, Nasifu Kerebba, Fanie Rautenbach, Sachin Kumar Singh, Kamal Dua, Oluwafemi O. Oguntibeju. UPLC-ESI-QTOF-MS phenolic compounds identification and quantification from ethanolic extract of Myrtus communis ‘Variegatha’: In vitro antioxidant and antidiabetic potentials. Arabian Journal of Chemistry 2023, 16 (2) , 104447. https://doi.org/10.1016/j.arabjc.2022.104447
    16. Xiao-Fang Song, Li-Jing Zhang, Xing-Guo Zhang, Hai-Yong Tu. Direct access to trifluoromethylated α-hydroxyketones from silver-catalyzed hydroacyloxylation of trifluoromethyl propynols with acids. Tetrahedron 2023, 130 , 133166. https://doi.org/10.1016/j.tet.2022.133166
    17. Feng Chen, Xiu-Hua Xu, Zeng-Hao Chen, Yue Chen, Feng-Ling Qing. Visible-light-induced nickel-catalyzed α-hydroxytrifluoroethylation of alkyl carboxylic acids: Access to trifluoromethyl alkyl acyloins. Beilstein Journal of Organic Chemistry 2023, 19 , 1372-1378. https://doi.org/10.3762/bjoc.19.98
    18. Kunle Okaiyeto, Nasifu Kerebba, Oluwafemi O. Oguntibeju. UPLC-ESI-QTOF-MS Profiling of Phenolic Compounds from Eriocephalus africanus: In Vitro Antioxidant, Antidiabetic, and Anti-Inflammatory Potentials. Molecules 2022, 27 (24) , 8912. https://doi.org/10.3390/molecules27248912
    19. Maryam Mohammadi‐Khanaposhtani, Milad Noori, Yousef Valizadeh, Navid Dastyafteh, Minoo Khalili Ghomi, Somayeh Mojtabavi, Mohammad Ali Faramarzi, Samanesadat Hosseini, Mahmood Biglar, Bagher Larijani, Hossein Rastegar, Haleh Hamedifar, Roghieh Mirzazadeh, Mohammad Mahdavi. Synthesis, α‐glucosidase Inhibition, in silico Pharmacokinetic, and Docking Studies Of Thieno[2,3‐b]Quinoline‐Acetamide Derivatives as New Anti‐Diabetic Agents. ChemistrySelect 2022, 7 (44) https://doi.org/10.1002/slct.202104482
    20. Milad Noori, Mryam Rastak, Mohammad Halimi, Minoo Khalili Ghomi, Mrjan Mollazadeh, Maryam Mohammadi-Khanaposhtani, Mohammad Hosein Sayahi, Zahra Rezaei, Somayeh Mojtabavi, Mohammad Ali Faramarzi, Bagher Larijani, Mahmood Biglar, Massoud Amanlou, Mohammad Mahdavi. Design, synthesis, in vitro, and in silico enzymatic evaluations of thieno[2,3-b]quinoline-hydrazones as novel inhibitors for α-glucosidase. Bioorganic Chemistry 2022, 127 , 105996. https://doi.org/10.1016/j.bioorg.2022.105996
    21. Tao Yang, Kailing Yang, Yu Zhang, Ruixi Zhou, Fuxin Zhang, Guanqun Zhan, Zengjun Guo. Metabolites with antioxidant and α -glucosidase inhibitory activities produced by the endophytic fungi Aspergillus niger from Pachysandra terminalis. Bioscience, Biotechnology, and Biochemistry 2022, 86 (10) , 1343-1348. https://doi.org/10.1093/bbb/zbac137
    22. Fuxin ZHANG, Rongkun MIAO, Kailing YANG, Tao YANG, Ruixi ZHOU, Guanqun ZHAN, Zengjun GUO. Two new phenylpropanoids from the leaves of Rauvolfia vomitoria. Journal of Holistic Integrative Pharmacy 2022, 3 (3) , 236-242. https://doi.org/10.1016/S2707-3688(23)00044-4
    23. Rachel Mata, Aldo J. Contreras-Rosales, José A. Gutiérrez-González, José L. Villaseñor, Araceli Pérez-Vásquez. Calea ternifolia Kunth, the Mexican “dream herb”, a concise review. Botany 2022, 100 (2) , 261-274. https://doi.org/10.1139/cjb-2021-0063
    24. R. Dhanya. Quercetin for managing type 2 diabetes and its complications, an insight into multitarget therapy. Biomedicine & Pharmacotherapy 2022, 146 , 112560. https://doi.org/10.1016/j.biopha.2021.112560
    25. Yeniley Ruiz-Noa, Lorena Del Rocío Ibarra-Reynoso, Alan Joel Ruiz-Padilla, Angel Josabad Alonso-Castro, Marco Antonio Ramírez-Morales, Juan Ramón Zapata-Morales, Luis Manuel Orozco-Castellanos, Cesar Rogelio Solorio-Alvardo, Alfredo Lara-Morales. Use of herbal medicine for diabetes mellitus in adults from the central–western region of Mexico. Primary Care Diabetes 2021, 15 (6) , 1095-1099. https://doi.org/10.1016/j.pcd.2021.08.010
    26. Jirawat Riyaphan, Dinh-Chuong Pham, Max K. Leong, Ching-Feng Weng. In Silico Approaches to Identify Polyphenol Compounds as α-Glucosidase and α-Amylase Inhibitors against Type-II Diabetes. Biomolecules 2021, 11 (12) , 1877. https://doi.org/10.3390/biom11121877
    27. Kai Yang, Feng Zhang, Tongchang Fang, Chaokun Li, Wangyang Li, Qiuling Song. Passerini-type reaction of boronic acids enables α-hydroxyketones synthesis. Nature Communications 2021, 12 (1) https://doi.org/10.1038/s41467-020-20727-7
    28. Mohibullah Shah, Sidra Bashir, Samavia Jaan, Haq Nawaz, Umar Nishan, Sumra Wajid Abbasi, Syed Babar Jamal, Asifullah Khan, Sahib Gul Afridi, Anwar Iqbal. Computational Analysis of Plant-Derived Terpenes as α -glucosidase Inhibitors for the Discovery of Therapeutic Agents against Type 2 Diabetes Mellitus. South African Journal of Botany 2021, 143 , 462-473. https://doi.org/10.1016/j.sajb.2021.09.030
    29. Fernando Calzada, Elihú Bautista, Sergio Hidalgo-Figueroa, Normand García-Hernández, Elizabeth Barbosa, Claudia Velázquez, Rosa María Ordoñez-Razo, Angel Giovanni Arietta-García. Antilymphoma Effect of Incomptine A: In Vivo, In Silico, and Toxicological Studies. Molecules 2021, 26 (21) , 6646. https://doi.org/10.3390/molecules26216646
    30. Adolfo Andrade-Cetto, Fernanda Espinoza-Hernández, Gerardo Mata-Torres, Sonia Escandón-Rivera. Hypoglycemic Effect of Two Mexican Medicinal Plants. Plants 2021, 10 (10) , 2060. https://doi.org/10.3390/plants10102060
    31. Fernanda Espinoza-Hernández, Adolfo Andrade-Cetto, Sonia Escandón-Rivera, Gerardo Mata-Torres, Rachel Mata. Contribution of fasting and postprandial glucose-lowering mechanisms to the acute hypoglycemic effect of traditionally used Eryngium cymosum F.Delaroche. Journal of Ethnopharmacology 2021, 279 , 114339. https://doi.org/10.1016/j.jep.2021.114339
    32. Elizabeth Barber, Michael J. Houghton, Gary Williamson. Flavonoids as Human Intestinal α-Glucosidase Inhibitors. Foods 2021, 10 (8) , 1939. https://doi.org/10.3390/foods10081939
    33. Virginia Gabriela Cilia-López, Raquel Cariño-Cortés, Luis Ricardo Zurita-Salinas. Ethnopharmacology of the Asteraceae family in Mexico. Botanical Sciences 2021, 99 (3) , 455-486. https://doi.org/10.17129/botsci.2715
    34. Liyao Ma, Yinghua Yu, Luoting Xin, Lei Zhu, Jiajin Xia, Pengcheng Ou, Xueliang Huang. Visible Light Enabled Formal Cross Silyl Benzoin Reaction as an Access to α‐Hydroxyketones. Advanced Synthesis & Catalysis 2021, 363 (10) , 2573-2577. https://doi.org/10.1002/adsc.202100186
    35. Manuel Jiménez-Estrada, Maira Huerta-Reyes, Rosario Tavera-Hernández, J. Javier Alvarado-Sansininea, Ana Berenice Alvarez. Contributions from Mexican Flora for the Treatment of Diabetes Mellitus: Molecules of Psacalium decompositum (A. Gray) H. Rob & Brettell. Molecules 2021, 26 (10) , 2892. https://doi.org/10.3390/molecules26102892
    36. Kesinee Nanok, Sompong Sansenya. Combination effects of rice extract and five aromatic compounds against α-glucosidase, α-amylase and tyrosinase. Journal of Bioscience and Bioengineering 2021, 2014 https://doi.org/10.1016/j.jbiosc.2021.02.003
    37. Yueqiu Liu, Wen Zheng, Yi Zhong, Lu Zhang, Tao Su, Ge Liang, Dingkun Zhang, Yong Zhang, Jingqiu Chen, Meng Gong. Identification of α-glucosidase inhibitors from Cortex Lycii based on a bioactivity-labeling high-resolution mass spectrometry–metabolomics investigation. Journal of Chromatography A 2021, 1642 , 462041. https://doi.org/10.1016/j.chroma.2021.462041
    38. Deependra Singh Chauhan, Paras Gupta, Faheem Hyder Pottoo, Mohd Amir. Secondary Metabolites in the Treatment of Diabetes Mellitus: A Paradigm Shift. Current Drug Metabolism 2020, 21 (7) , 493-511. https://doi.org/10.2174/1389200221666200514081947
    39. Shilpi Singh, Pratima Gupta, Abha Meena, Suaib Luqman. Acacetin, a flavone with diverse therapeutic potential in cancer, inflammation, infections and other metabolic disorders. Food and Chemical Toxicology 2020, 145 , 111708. https://doi.org/10.1016/j.fct.2020.111708
    40. Sonia Marlen Escandón-Rivera, Rachel Mata, Adolfo Andrade-Cetto. Molecules Isolated from Mexican Hypoglycemic Plants: A Review. Molecules 2020, 25 (18) , 4145. https://doi.org/10.3390/molecules25184145
    41. Sompong Sansenya, Kesinee Nanok. α‐glucosidase, α‐amylase inhibitory potential and antioxidant activity of fragrant black rice (Thai coloured rice). Flavour and Fragrance Journal 2020, 35 (4) , 376-386. https://doi.org/10.1002/ffj.3572
    42. Bathini Thissera, Rizliya Visvanathan, Mohammad A. Khanfar, M. Mallique Qader, Marwa H.A. Hassan, Hossam M. Hassan, Majed Bawazeer, Fathy A. Behery, Mohammed Yaseen, Ruvini Liyanage, Usama R. Abdelmohsen, Mostafa E. Rateb. Sesbania grandiflora L. Poir leaves: A dietary supplement to alleviate type 2 diabetes through metabolic enzymes inhibition. South African Journal of Botany 2020, 130 , 282-299. https://doi.org/10.1016/j.sajb.2020.01.011
    43. Kang Chen, Xiao-Qin Liu, Wen-Li Wang, Jian-Guang Luo, Ling-Yi Kong. Taxumarienes A–G, seven new α-glucosidase inhibitory taxane-diterpenoids from the leaves of Taxus mairei. Bioorganic Chemistry 2020, 94 , 103400. https://doi.org/10.1016/j.bioorg.2019.103400
    44. Gabriela López-Angulo, Julio Montes-Avila, Sylvia Páz Díaz-Camacho, Rito Vega-Aviña, Yesmi Patricia Ahumada-Santos, Francisco Delgado-Vargas. Chemical composition and antioxidant, α-glucosidase inhibitory and antibacterial activities of three Echeveria DC. species from Mexico. Arabian Journal of Chemistry 2019, 12 (8) , 1964-1973. https://doi.org/10.1016/j.arabjc.2014.11.050
    45. Mohamad Fawzi Mahomoodally, Gokhan Zengin, Mustafa Onur Aladag, Haluk Ozparlak, Alina Diuzheva, József Jekő, Zoltán Cziáky, Muhammad Zakariyyah Aumeeruddy. HPLC-MS/MS chemical characterization and biological properties of Origanum onites extracts: a recent insight. International Journal of Environmental Health Research 2019, 29 (6) , 607-621. https://doi.org/10.1080/09603123.2018.1558184
    46. Fatih Yılmaz, Emre Menteşe, Nimet Baltaş. Synthesis and Biological Evaluation of Some Succinimide Hybrid Molecules. Russian Journal of Bioorganic Chemistry 2019, 45 (6) , 575-584. https://doi.org/10.1134/S1068162019060402
    47. Rui Li, Qing Wang, Menghao Zhao, Peiming Yang, Xiao Hu, Danwei Ouyang. Flavonoid glycosides from seeds of Hippophae rhamnoides subsp. Sinensis with α-glucosidase inhibition activity. Fitoterapia 2019, 137 , 104248. https://doi.org/10.1016/j.fitote.2019.104248
    48. T.S. Rizvi, I. Hussain, L. Ali, F. Mabood, A.L. Khan, S. Shujah, N.U. Rehman, A. Al-Harrasi, J. Hussain, A. Khan, S.A. Halim. New gorgonane sesquiterpenoid from Teucrium mascatense Boiss, as α-glucosidase inhibitor. South African Journal of Botany 2019, 124 , 218-222. https://doi.org/10.1016/j.sajb.2019.05.008
    49. Liangjin Xu, Tao Huang, Chunyue Huang, Chunzhen Wu, An Jia, Xiao Hu. Chiral separation, absolute configuration, and bioactivity of two pairs of flavonoid enantiomers from Morus nigra. Phytochemistry 2019, 163 , 33-37. https://doi.org/10.1016/j.phytochem.2019.03.029
    50. Bhausaheb N. Patil, Jatin J. Lade, Aniket A. Parab, Pratima A. Sathe, Kamlesh S. Vadagaonkar, Atul C. Chaskar. NBS-assisted an efficient conversion of styrenes to α-hydroxy ketones in water. Tetrahedron Letters 2019, 60 (27) , 1788-1791. https://doi.org/10.1016/j.tetlet.2019.06.004
    51. Lei Chen, Xu Lu, Hesham El-Seedi, Hui Teng. Recent advances in the development of sesquiterpenoids in the treatment of type 2 diabetes. Trends in Food Science & Technology 2019, 88 , 46-56. https://doi.org/10.1016/j.tifs.2019.02.003
    52. Junghyun Kim, Soo-Wang Hyun, Ik Soo Lee, Kyuhyung Jo, Young Sook Kim, Jin Sook Kim, Chan-Sik Kim. Aster koraiensis extract lowers postprandial glucose in normoglycemic and high-fat-diet-induced obese mice. Food Science and Biotechnology 2019, 28 (2) , 563-568. https://doi.org/10.1007/s10068-018-0497-8
    53. Xin Cheng, Yan Huang, Chao Yuan, Ke Dai, Hong Jiang, Jimei Ma. Colorimetric detection of α-glucosidase activity based on the etching of gold nanorods and its application to screen anti-diabetic drugs. Sensors and Actuators B: Chemical 2019, 282 , 838-843. https://doi.org/10.1016/j.snb.2018.11.097
    54. Rachel Mata, Mario Figueroa, Andrés Navarrete, Isabel Rivero-Cruz. Chemistry and Biology of Selected Mexican Medicinal Plants. 2019, 1-142. https://doi.org/10.1007/978-3-030-01099-7_1
    55. Zhen-Tao Deng, Chang-An Geng, Tong-Hua Yang, Chun-Lei Xiang, Ji-Jun Chen. Chepraecoxins A-G, ent-Kaurane Diterpenoids with α-Glucosidase Inhibitory Activities from Chelonopsis praecox. Fitoterapia 2019, 132 , 60-67. https://doi.org/10.1016/j.fitote.2018.11.014
    56. Hajer Ben Saad, Ibtissem Ben Amara, Nadia Kharrat, Marie-Agnès Giroux-Metgès, Ahmed Hakim, Khaled Mounir Zeghal, Hélène Talarmin. Cytoprotective and antioxidant effects of the red alga Alsidium corallinum against hydrogen peroxide-induced toxicity in rat cardiomyocytes. Archives of Physiology and Biochemistry 2019, 125 (1) , 35-43. https://doi.org/10.1080/13813455.2018.1437184
    57. Laura Flores-Bocanegra, Rafael Torres-Colín, Martin González-Andrade, José S. Calderón, Rachel Mata. In Vivo and In Vitro α-Glucosidase Inhibitory Activity of Perfoliatin a from Melampodium Perfoliatum. Natural Product Communications 2019, 14 (1) , 1934578X1901400. https://doi.org/10.1177/1934578X1901400102
    58. Daniel Rosas-Ramírez, Sonia Escandón-Rivera, Rogelio Pereda-Miranda. Morning glory resin glycosides as α-glucosidase inhibitors: In vitro and in silico analysis. Phytochemistry 2018, 148 , 39-47. https://doi.org/10.1016/j.phytochem.2018.01.012
    59. . 2,2‐Dimethylbenzopyrans. 2018, 414-434. https://doi.org/10.1002/9781118686263.ch11
    60. Fan Xie, Shaoyun Wang, Li Zhang, Jinhong Wu, Zhengwu Wang. Investigating inhibitory activity of novel synthetic sericin peptide on α ‐D‐glucosidase: kinetics and interaction mechanism study using a docking simulation. Journal of the Science of Food and Agriculture 2018, 98 (4) , 1502-1510. https://doi.org/10.1002/jsfa.8620
    61. Janggyoo Choi, Kee Dong Yoon, Jinwoong Kim. Chemical constituents from Taraxacum officinale and their α-glucosidase inhibitory activities. Bioorganic & Medicinal Chemistry Letters 2018, 28 (3) , 476-481. https://doi.org/10.1016/j.bmcl.2017.12.014
    62. Etetor Roland Eshiet, Ernest E. Smith. Herbal Benefits of Tea. 2018, 287-320. https://doi.org/10.4018/978-1-5225-5207-9.ch014
    63. Cong Tang, Zhaosheng Qian, Yinjie Qian, Yuanyuan Huang, Meizhi Zhao, Hang Ao, Hui Feng. A fluorometric and real-time assay for α-glucosidase activity through supramolecular self-assembly and its application for inhibitor screening. Sensors and Actuators B: Chemical 2017, 245 , 282-289. https://doi.org/10.1016/j.snb.2017.01.150
    64. Fan Xie, Shengxiang Gong, Wei Zhang, Jinhong Wu, Zhengwu Wang. Potential of lignin from Canna edulis ker residue in the inhibition of α-d-glucosidase: Kinetics and interaction mechanism merging with docking simulation. International Journal of Biological Macromolecules 2017, 95 , 592-602. https://doi.org/10.1016/j.ijbiomac.2016.11.100
    65. Sonia Escandón-Rivera, Araceli Pérez-Vásquez, Andrés Navarrete, Mariana Hernández, Edelmira Linares, Robert Bye, Rachel Mata. Anti-Hyperglycemic Activity of Major Compounds from Calea ternifolia. Molecules 2017, 22 (2) , 289. https://doi.org/10.3390/molecules22020289
    66. D. Chaturvedi, P.K. Dwivedi. Recent Developments on the Antidiabetic Sesquiterpene Lactones and Their Semisynthetic Analogues. 2017, 185-207. https://doi.org/10.1016/B978-0-12-809450-1.00006-5
    67. Dong Young Lee, Heejung Yang, Hyun Woo Kim, Sang Hyun Sung. New polyhydroxytriterpenoid derivatives from fruits of Terminalia chebula Retz. and their α-glucosidase and α-amylase inhibitory activity. Bioorganic & Medicinal Chemistry Letters 2017, 27 (1) , 34-39. https://doi.org/10.1016/j.bmcl.2016.11.039
    68. Etetor Roland Eshiet, Ernest E. Smith. Herbal Benefits of Tea. 2017, 117-146. https://doi.org/10.4018/978-1-5225-0591-4.ch006
    69. Gabriela Ávila-Villarreal, María Eva González-Trujano, Azucena Ibeth Carballo-Villalobos, Berenice Aguilar-Guadarrama, Sara García-Jiménez, Diana Elizabeth Giles-Rivas, Patricia Castillo-España, Rafael Villalobos-Molina, Samuel Estrada-Soto. Anxiolytic-like effects and toxicological studies of Brickellia cavanillesii (Cass.) A. Gray in experimental mice models. Journal of Ethnopharmacology 2016, 192 , 90-98. https://doi.org/10.1016/j.jep.2016.07.006
    70. Si‐Yao Liu, Huan Wang, Tian He, Liang Qi, Zhi‐Qi Zhang. Sensitive fluorimetric assays for α‐glucosidase activity and inhibitor screening based on β‐cyclodextrin‐coated quantum dots. Luminescence 2016, 31 (1) , 96-101. https://doi.org/10.1002/bio.2929
    71. Xi Peng, Guowen Zhang, Yijing Liao, Deming Gong. Inhibitory kinetics and mechanism of kaempferol on α-glucosidase. Food Chemistry 2016, 190 , 207-215. https://doi.org/10.1016/j.foodchem.2015.05.088
    72. Li Zeng, Guowen Zhang, Yijing Liao, Deming Gong. Inhibitory mechanism of morin on α-glucosidase and its anti-glycation properties. Food & Function 2016, 7 (9) , 3953-3963. https://doi.org/10.1039/C6FO00680A
    73. Bin Liu, Ji-Mei Ma, Hang-Wei Chen, Zi-Long Li, Lin-Hao Sun, Zhen Zeng, Hong Jiang. α-Glucosidase inhibitory activities of phenolic acid amides with l -amino acid moiety. RSC Advances 2016, 6 (56) , 50837-50845. https://doi.org/10.1039/C6RA08330G
    74. Ya‐Long Zhang, Jian‐Guang Luo, Chuan‐Xing Wan, Zhong‐Bo Zhou, Ling‐Yi Kong. Four New Flavonoids with α ‐Glucosidase Inhibitory Activities from Morus alba var. tatarica. Chemistry & Biodiversity 2015, 12 (11) , 1768-1776. https://doi.org/10.1002/cbdv.201500005
    75. Malek Besbes Hlila, Habib Mosbah, Kawther Majouli, Kamel Msaada, Hichem Ben Jannet, Mahjoub Aouni, Boulbaba Selmi. α-Glucosidase inhibition by Tunisian Scabiosa arenaria Forssk. extracts. International Journal of Biological Macromolecules 2015, 77 , 383-389. https://doi.org/10.1016/j.ijbiomac.2015.03.035
    76. Edward E. Schilling, Jose L. Panero, Bonnie S. Crozier, Randall W. Scott, Patricia Dávila. Bricklebush (Brickellia) phylogeny reveals dimensions of the great Asteraceae radiation in Mexico. Molecular Phylogenetics and Evolution 2015, 85 , 161-170. https://doi.org/10.1016/j.ympev.2015.02.007
    77. You Yang, Gaoyan Lian, Biao Yu. Naturally Occurring Polyphenolic Glucosidase Inhibitors. Israel Journal of Chemistry 2015, 55 (3-4) , 268-284. https://doi.org/10.1002/ijch.201400134
    78. Krutzkaya Juárez-Reyes, Fernando Brindis, Omar N. Medina-Campos, José Pedraza-Chaverri, Robert Bye, Edelmira Linares, Rachel Mata. Hypoglycemic, antihyperglycemic, and antioxidant effects of the edible plant Anoda cristata. Journal of Ethnopharmacology 2015, 161 , 36-45. https://doi.org/10.1016/j.jep.2014.11.052
    79. Jian Chen, Sven Mangelinckx, An Adams, Zheng-tao Wang, Wei-lin Li, Norbert De Kimpe. Natural Flavonoids as Potential Herbal Medication for the Treatment of Diabetes Mellitus and its Complications. Natural Product Communications 2015, 10 (1) , 1934578X1501000. https://doi.org/10.1177/1934578X1501000140
    80. Hui-Yu Fang, Uvarani Chokkalingam, Shu-Fen Chiou, Tsong-Long Hwang, Shu-Li Chen, Wei-Lung Wang, Jyh-Horng Sheu. Bioactive Chemical Constituents from the Brown Alga Homoeostrichus formosana. International Journal of Molecular Sciences 2015, 16 (1) , 736-746. https://doi.org/10.3390/ijms16010736
    81. Shi-Mei Wang, Jun-Jie Han, Ke Ma, Tao Jin, Li Bao, Yun-Fei Pei, Hong-Wei Liu. New α-glucosidase inhibitors with p-terphenyl skeleton from the mushroom Hydnellum concrescens. Fitoterapia 2014, 98 , 149-155. https://doi.org/10.1016/j.fitote.2014.07.019
    82. Zhenhua Yin, Wei Zhang, Fajin Feng, Yong Zhang, Wenyi Kang. α-Glucosidase inhibitors isolated from medicinal plants. Food Science and Human Wellness 2014, 3 (3-4) , 136-174. https://doi.org/10.1016/j.fshw.2014.11.003
    83. Dongsoo Koh. Crystal structure of 1-(8-methoxy-2 H -chromen-3-yl)ethanone. Acta Crystallographica Section E Structure Reports Online 2014, 70 (9) , o936-o937. https://doi.org/10.1107/S1600536814016808
    84. Gerardo D. Anaya-Eugenio, Isabel Rivero-Cruz, José Rivera-Chávez, Rachel Mata. Hypoglycemic properties of some preparations and compounds from Artemisia ludoviciana Nutt. Journal of Ethnopharmacology 2014, 155 (1) , 416-425. https://doi.org/10.1016/j.jep.2014.05.051
    85. Jiakai Yan, Guowen Zhang, Junhui Pan, Yajie Wang. α-Glucosidase inhibition by luteolin: Kinetics, interaction and molecular docking. International Journal of Biological Macromolecules 2014, 64 , 213-223. https://doi.org/10.1016/j.ijbiomac.2013.12.007
    86. Ya-Long Zhang, Jian-Guang Luo, Chuan-Xing Wan, Zhong-Bo Zhou, Ling-Yi Kong. Geranylated 2-arylbenzofurans from Morus alba var. tatarica and their α-glucosidase and protein tyrosine phosphatase 1B inhibitory activities. Fitoterapia 2014, 92 , 116-126. https://doi.org/10.1016/j.fitote.2013.10.017
    87. Ana Laura de la Garza, Usune Etxeberria, Marı́a Pilar Lostao, Belén San Román, Jaione Barrenetxe, J. Alfredo Martínez, Fermı́n I. Milagro. Helichrysum and Grapefruit Extracts Inhibit Carbohydrate Digestion and Absorption, Improving Postprandial Glucose Levels and Hyperinsulinemia in Rats. Journal of Agricultural and Food Chemistry 2013, 61 (49) , 12012-12019. https://doi.org/10.1021/jf4021569
    88. Chunfang Zhao, Yongqiang Liu, Dengli Cong, Hui Zhang, Jiajia Yu, Ying Jiang, Xiuyue Cui, Jiaming Sun. Screening and determination for potential α ‐glucosidase inhibitory constituents from Dalbergia odorifera T. Chen using ultrafiltration‐LC/ESI‐MS n . Biomedical Chromatography 2013, 27 (12) , 1621-1629. https://doi.org/10.1002/bmc.2970
    89. José Rivera-Chávez, Martín González-Andrade, María del Carmen González, Anthony E. Glenn, Rachel Mata. Thielavins A, J and K: α-Glucosidase inhibitors from MEXU 27095, an endophytic fungus from Hintonia latiflora. Phytochemistry 2013, 94 , 198-205. https://doi.org/10.1016/j.phytochem.2013.05.021
    90. Hongyu Wang, Tingting Liu, Dejian Huang. Starch Hydrolase Inhibitors from Edible Plants. 2013, 103-136. https://doi.org/10.1016/B978-0-12-416555-7.00003-5
    91. Dongsoo Koh. 8-Methoxy-2 H -chromene-3-carbaldehyde. Acta Crystallographica Section E Structure Reports Online 2012, 68 (12) , o3419-o3419. https://doi.org/10.1107/S1600536812047319

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    You’ve supercharged your research process with ACS and Mendeley!

    STEP 1:
    Click to create an ACS ID

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    MENDELEY PAIRING EXPIRED
    Your Mendeley pairing has expired. Please reconnect