1
|
Kazempour-Dizaji M, Mojtabavi S, Sadri A, Ghanbarpour A, Faramarzi MA, Navidpour L. Arylureidoaurones: Synthesis, in vitro α-glucosidase, and α-amylase inhibition activity. Bioorg Chem 2023; 139:106709. [PMID: 37442042 DOI: 10.1016/j.bioorg.2023.106709] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
Because of the colossal global burden of diabetes, there is an urgent need for more effective and safer drugs. We designed and synthesized a new series of aurone derivatives possessing phenylureido or bis-phenylureido moieties as α-glucosidase and α-amylase inhibitors. Most of the synthesized phenylureidoaurones have demonstrated superior inhibition activities (IC50s of 9.6-339.9 μM) against α-glucosidase relative to acarbose (IC50 = 750.0 μM) as the reference drug. Substitution of aurone analogues with two phenylureido substituents at the 5-position of the benzofuranone moiety and the 3' or 4' positions of the 2-phenyl ring resulted in compounds with almost 120-180 times more potent inhibitory activities than acarbose. The aurone analogue possessing two phenylureido substitutions at 5 and 4' positions (13) showed the highest inhibition activity with an IC50 of 4.2 ± 0.1 μM. Kinetic studies suggested their inhibition mode to be competitive. We also investigated the binding mode of the most potent compounds using the consensually docked 4D-QSAR methodology. Furthermore, these analogues showed weak-to-moderate non-competitive inhibitory activity against α-amylase. 5-Methyl substituted aurone with 4'-phenylureido moiety (6e) demonstrated the highest inhibition activity on α-amylase with an IC50 of 142.0 ± 1.6 μM relative to acarbose (IC50 = 108 ± 1.2 μM). Our computational studies suggested that these analogues interact with a hydrophilic allosteric site in α-amylase, located far from the enzyme active site at the N-terminal.
Collapse
Affiliation(s)
- Mohammad Kazempour-Dizaji
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 14176, Iran
| | - Arash Sadri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran; Interdisciplinary Neuroscience Research Program, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran; Lyceum Scientific Charity, Iran
| | - Araz Ghanbarpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 14176, Iran
| | - Latifeh Navidpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran.
| |
Collapse
|
2
|
Pasha AR, Khan A, Ullah S, Halim SA, Hussain J, Khalid M, Naseer MM, El-Kott AF, Negm S, Al-Harrasi A, Shafiq Z. Synthesis of new diphenyl urea-clubbed imine analogs and its Implications in diabetic management through in vitro and in silico approaches. Sci Rep 2023; 13:1877. [PMID: 36725861 PMCID: PMC9892044 DOI: 10.1038/s41598-023-28828-1] [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: 10/19/2022] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Type II diabetes mellitus (T2DM) is a global health issue with high rate of prevalence. The inhibition of α-glucosidase enzyme has prime importance in the management of T2DM. This study was established to synthesize Schiff bases of 1,3-dipheny urea (3a-y) and to investigate their in vitro anti-diabetic capability via inhibiting α-glucosidase, a key player in the catabolism of carbohydrates. The structures of all compounds were confirmed through various techniques including, Fourier-transform infrared spectroscopy (FTIR) and nuclear magnetic resonance (NMR) and mass-spectrometry (MS) methods. Interestingly all these compounds displayed potent inhibition IC50 values in range of 2.14-115 µM as compared to acarbose used as control. Additionally, all the compounds were docked at the active site of α-glucosidase to predict their mode of binding. The docking results indicates that Glu277 and Asn350 play important role in the stabilization of these compounds in the active site of enzyme. These molecules showed excellent predicted pharmacokinetics, physicochemical and drug-likeness profile. The anti-diabetic potential of these molecules signifies their medical importance and provide insights into prospective therapeutic options for the treatment of T2DM.
Collapse
Affiliation(s)
- Anam Rubbab Pasha
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan.,Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman.,International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Javid Hussain
- Department of Biological Sciences and Chemistry, University of Nizwa, Nizwa-616, Nizwa, Oman
| | - Muhammad Khalid
- Department of Chemistry, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, 64200, Pakistan.,Centre for Theoretical and Computational Research, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, 64200, Pakistan
| | | | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, 61421, Abha, Saudi Arabia.,Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Sally Negm
- Department of Life Sciences, College of Science and Art Mahyel Aseer, King Khalid University, 62529, Abha, Saudi Arabia.,Unit of Food Bacteriology, Central Laboratory of Food Hygiene, Ministry of Health, Branch in Zagazig, Zagazig, 44511, Egypt
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman.
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan. .,Department of Pharmaceutical and Medicinal Chemistry, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
3
|
Baumer KM, Cook CD, Zahler CT, Beard AA, Chen Z, Koone JC, Dashnaw CM, Villacob RA, Solouki T, Wood JL, Borchelt DR, Shaw BF. Supercharging Prions via Amyloid‐Selective Lysine Acetylation. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Katelyn M. Baumer
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | | | - Collin T. Zahler
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | | | - Zhijuan Chen
- Department of Neuroscience University of Florida Gainesville FL USA
| | - Jordan C. Koone
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Chad M. Dashnaw
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Raul A. Villacob
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Touradj Solouki
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - John L. Wood
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | | | - Bryan F. Shaw
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| |
Collapse
|
4
|
Baumer KM, Cook CD, Zahler CT, Beard AA, Chen Z, Koone JC, Dashnaw CM, Villacob RA, Solouki T, Wood JL, Borchelt DR, Shaw BF. Supercharging Prions via Amyloid-Selective Lysine Acetylation. Angew Chem Int Ed Engl 2021; 60:15069-15079. [PMID: 33876528 DOI: 10.1002/anie.202103548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Indexed: 11/10/2022]
Abstract
Repulsive electrostatic forces between prion-like proteins are a barrier against aggregation. In neuropharmacology, however, a prion's net charge (Z) is not a targeted parameter. Compounds that selectively boost prion Z remain unreported. Here, we synthesized compounds that amplified the negative charge of misfolded superoxide dismutase-1 (SOD1) by acetylating lysine-NH3 + in amyloid-SOD1, without acetylating native-SOD1. Compounds resembled a "ball and chain" mace: a rigid amyloid-binding "handle" (benzothiazole, stilbene, or styrylpyridine); an aryl ester "ball"; and a triethylene glycol chain connecting ball to handle. At stoichiometric excess, compounds acetylated up to 9 of 11 lysine per misfolded subunit (ΔZfibril =-8100 per 103 subunits). Acetylated amyloid-SOD1 seeded aggregation more slowly than unacetylated amyloid-SOD1 in vitro and organotypic spinal cord (these effects were partially due to compound binding). Compounds exhibited reactivity with other amyloid and non-amyloid proteins (e.g., fibrillar α-synuclein was peracetylated; serum albumin was partially acetylated; carbonic anhydrase was largely unacetylated).
Collapse
Affiliation(s)
- Katelyn M Baumer
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Christopher D Cook
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Collin T Zahler
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Alexandra A Beard
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Zhijuan Chen
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Jordan C Koone
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Chad M Dashnaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Raul A Villacob
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Touradj Solouki
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - John L Wood
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - David R Borchelt
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Bryan F Shaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| |
Collapse
|
5
|
Anti-insulin resistance effect of constituents from Senna siamea on zebrafish model, its molecular docking, and structure-activity relationships. J Nat Med 2021; 75:520-531. [PMID: 33620670 DOI: 10.1007/s11418-021-01490-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/20/2021] [Indexed: 01/24/2023]
Abstract
Senna siamea has been used as an antidiabetic drug since antiquity. With regard to traditional Thai medicine, the use of S. siamea was described for diabetes therapy. To understand the molecular mechanism regarding insulin resistance. Pure compounds were isolated from wood extract. We studied their biological activities on insulin-resistance using an in vivo zebrafish model. We also performed an in silico study; molecular docking, and in vitro study by taking advantage of the enzyme inhibitory activities of α-glucosidase, PTP1B, and DPP-IV. Based on the preliminary investigation that ethyl acetate and ethanol extracts have potent effects against insulin resistance on zebrafish larvae, five compounds were isolated from two fractions following: resveratrol, piceatannol, dihydropiceatannol, chrysophanol, and emodin. All of the isolated compounds had anti-insulin resistance effects on zebrafish larvae. Resveratrol, piceatannol, and dihydropiceatannol also demonstrated inhibitory effects against α-glucosidase. Chrysophanol and emodin inhibited PTP1B activity, while resveratrol showed a DPP-IV inhibition effect via the molecular docking. The results of enzyme assay were similar. In conclusions, S. siamea components demonstrated effects against insulin resistance. The chemical structure displayed identical biological activity to that of the compounds. Therefore, S. siamea wood extract and their components are potential therapeutic options in the treatment of diabetes.
Collapse
|
6
|
Jenis J, Baiseitova A, Yoon SH, Park C, Kim JY, Li ZP, Lee KW, Park KH. Competitive α-glucosidase inhibitors, dihydrobenzoxanthones, from the barks of Artocarpus elasticus. J Enzyme Inhib Med Chem 2020; 34:1623-1632. [PMID: 31480857 PMCID: PMC6735331 DOI: 10.1080/14756366.2019.1660653] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This study aimed to search the α-glucosidase inhibitors from the barks part of
Artocarpus elasticus. The responsible compounds for α-glucosidase
inhibition were found out as dihydrobenzoxanthones (1–4) and
alkylated flavones (5–6). All compounds showed a significant
enzyme inhibition toward α-glucosidase with IC50s of 7.6–25.4 μM.
Dihydrobenzoxanthones (1–4) exhibited a competitive inhibition
to α-glucosidase. This competitive behaviour was fully characterised by double reciprocal
plots, Yang’s method, and time-dependent experiments. The compound 1
manifested as the competitive and reversible simple slow-binding, with kinetic parameters
k3 = 0.0437 µM−1 min−1,
k4 = 0.0166 min−1, and Kiapp=
0.3795 µM. Alkylated flavones (5–6) were mixed type I
(KI < KIS) inhibitors. The
binding affinities (KSV) represented by all inhibitors were
correlated to their concentrations and inhibitory potencies (IC50). Moreover,
compounds 1 and 5 were identified as new ones named as
artoindonesianin W and artoflavone B, respectively. Molecular modelling study proposed the
putative binding conformation of competitive inhibitors (1–4) to
α-glucosidase at the atomic level.
Collapse
Affiliation(s)
- Janar Jenis
- Research Center for Medicinal Plants, Al-Farabi Kazakh National University , Almaty , Kazakhstan
| | - Aizhamal Baiseitova
- Division of Applied Life Science (BK21 plus), IALS, Gyeongsang National University , Jinju , Republic of Korea
| | - Sang Hwa Yoon
- Division of Applied Life Science (BK21 plus), PMBBRC, RINS, Gyeongsang National University , Jinju , Republic of Korea
| | - Chanin Park
- Division of Applied Life Science (BK21 plus), PMBBRC, RINS, Gyeongsang National University , Jinju , Republic of Korea
| | - Jeong Yoon Kim
- Division of Applied Life Science (BK21 plus), IALS, Gyeongsang National University , Jinju , Republic of Korea
| | - Zuo Peng Li
- Division of Applied Life Science (BK21 plus), IALS, Gyeongsang National University , Jinju , Republic of Korea
| | - Keun Woo Lee
- Division of Applied Life Science (BK21 plus), PMBBRC, RINS, Gyeongsang National University , Jinju , Republic of Korea
| | - Ki Hun Park
- Division of Applied Life Science (BK21 plus), IALS, Gyeongsang National University , Jinju , Republic of Korea
| |
Collapse
|
7
|
|
8
|
Mohd Bukhari DA, Siddiqui MJ, Shamsudin SH, Rahman MM, So'ad SZM. α-Glucosidase Inhibitory Activity of Selected Malaysian Plants. J Pharm Bioallied Sci 2017; 9:164-170. [PMID: 28979070 PMCID: PMC5621178 DOI: 10.4103/jpbs.jpbs_35_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Diabetes is a common metabolic disease indicated by unusually high plasma glucose level that can lead to major complications such as diabetic neuropathy, retinopathy, and cardiovascular diseases. One of the effective therapeutic managements of the disease is to reduce postprandial hyperglycemia through inhibition of α-glucosidase, a carbohydrate-hydrolyzing enzyme to retard overall glucose absorption. In recent years, a plenty of research works have been conducted looking for novel and effective α-glucosidase inhibitors (AGIs) from natural sources as alternatives for the synthetic AGI due to their unpleasant side effects. Plants and herbs are rich with secondary metabolites that have massive pharmaceutical potential. Besides, studies showed that phytochemicals such as flavonoids, alkaloids, terpenoids, anthocyanins, glycosides, and phenolic compounds possess significant inhibitory activity against α-glucosidase enzyme. Malaysia is a tropical country that is rich with medicinal herbs. In this review, we focus on eight Malaysian plants with the potential as AGI to develop a potential functional food or lead compounds against diabetes.
Collapse
Affiliation(s)
- Dzatil Awanis Mohd Bukhari
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Mohammad Jamshed Siddiqui
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Siti Hadijah Shamsudin
- Department of Pharmacy Practice, Kulliyyah of Pharmacy, International Islamic University Malaysia, Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Md Mukhlesur Rahman
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Siti Zaiton Mat So'ad
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| |
Collapse
|
9
|
Ghani U. Re-exploring promising α-glucosidase inhibitors for potential development into oral anti-diabetic drugs: Finding needle in the haystack. Eur J Med Chem 2015; 103:133-62. [PMID: 26344912 DOI: 10.1016/j.ejmech.2015.08.043] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 08/16/2015] [Accepted: 08/23/2015] [Indexed: 01/27/2023]
Abstract
Treatment of diabetes mellitus by oral α-glucosidase inhibitors is currently confined to acarbose, miglitol and voglibose marred by efficacy problems and unwanted side effects. Since the discovery of the drugs more than three decades ago, no significant progress has been made in the drug development area of anti-diabetic α-glucosidase inhibitors. Despite existence of a wide chemical diversity of α-glucosidase inhibitors identified to date, majority of them are simply piled up in publications and reports thus creating a haystack destined to be forgotten in the scientific literature without given consideration for further development into drugs. This review finds those "needles" in that haystack and lays groundwork for highlighting promising α-glucosidase inhibitors from the literature that may potentially become suitable candidates for pre-clinical or clinical trials while drawing attention of the drug development community to consider and take already-identified promising α-glucosidase inhibitors into the next stage of drug development.
Collapse
Affiliation(s)
- Usman Ghani
- Clinical Chemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia.
| |
Collapse
|
10
|
Anti-hyperglycemic activity of polyphenols isolated from barnyard millet (Echinochloa utilis L.) and their role inhibiting α-glucosidase. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s13765-015-0070-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
11
|
Lee Y, Kim S, Kim JY, Arooj M, Kim S, Hwang S, Kim BW, Park KH, Lee KW. Binding mode analyses and pharmacophore model development for stilbene derivatives as a novel and competitive class of α-glucosidase inhibitors. PLoS One 2014; 9:e85827. [PMID: 24465730 PMCID: PMC3897524 DOI: 10.1371/journal.pone.0085827] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 12/02/2013] [Indexed: 11/18/2022] Open
Abstract
Stilbene urea derivatives as a novel and competitive class of non-glycosidic α-glucosidase inhibitors are effective for the treatment of type II diabetes and obesity. The main purposes of our molecular modeling study are to explore the most suitable binding poses of stilbene derivatives with analyzing the binding affinity differences and finally to develop a pharmacophore model which would represents critical features responsible for α-glucosidase inhibitory activity. Three-dimensional structure of S. cerevisiae α-glucosidase was built by homology modeling method and the structure was used for the molecular docking study to find out the initial binding mode of compound 12, which is the most highly active one. The initial structure was subjected to molecular dynamics (MD) simulations for protein structure adjustment at compound 12-bound state. Based on the adjusted conformation, the more reasonable binding modes of the stilbene urea derivatives were obtained from molecular docking and MD simulations. The binding mode of the derivatives was validated by correlation analysis between experimental Ki value and interaction energy. Our results revealed that the binding modes of the potent inhibitors were engaged with important hydrogen bond, hydrophobic, and π-interactions. With the validated compound 12-bound structure obtained from combining approach of docking and MD simulation, a proper four featured pharmacophore model was generated. It was also validated by comparison of fit values with the Ki values. Thus, these results will be helpful for understanding the relationship between binding mode and bioactivity and for designing better inhibitors from stilbene derivatives.
Collapse
Affiliation(s)
- Yuno Lee
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Songmi Kim
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Jun Young Kim
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Mahreen Arooj
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Siu Kim
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Swan Hwang
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Byeong-Woo Kim
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Ki Hun Park
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Keun Woo Lee
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| |
Collapse
|
12
|
Zheng J, Li X, Wu H, Yan S, Ma L. WITHDRAWN: Polyhydroxybenzaldehyde azines: A novel competitive α-glucosidase inhibitor. Bioorg Med Chem 2013. [DOI: 10.1016/j.bmc.2013.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Kashima Y, Yamaki H, Suzuki T, Miyazawa M. Structure-activity relationships of bergenin derivatives effect on α-glucosidase inhibition. J Enzyme Inhib Med Chem 2012; 28:1162-70. [DOI: 10.3109/14756366.2012.719503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yusei Kashima
- Department of Applied Chemistry, Faculty of Science and Engineering, Kinki University,
Kowakae, Higashiosaka-shi, Osaka, Japan
| | - Hidehiko Yamaki
- Koei Kogyo Co., Ltd.,
Kanda-Awajicho, Chiyoda-ku, Tokyo, Japan
| | - Takuya Suzuki
- Koei Kogyo Co., Ltd.,
Kanda-Awajicho, Chiyoda-ku, Tokyo, Japan
| | - Mitsuo Miyazawa
- Department of Applied Chemistry, Faculty of Science and Engineering, Kinki University,
Kowakae, Higashiosaka-shi, Osaka, Japan
| |
Collapse
|
14
|
Ryu HW, Jeong SH, Curtis-Long MJ, Jung S, Lee JW, Woo HS, Cho JK, Park KH. Inhibition effects of mangosenone F from Garcinia mangostana on melanin formation in B16F10 cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:8372-8378. [PMID: 22779928 DOI: 10.1021/jf3015987] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Melanogenesis can be controlled by tyrosinase inhibition or by blocking the maturation processes of tyrosinase and its related proteins. Mangostenone F was isolated from the seedcases of Garcinia mangostana . Mangostenone F was shown to be inactive against tyrosinase (IC50 > 200 μM) but was a potent α-glucosidase inhibitor in vitro (IC50 = 21.0 μM). Mangostenone F was found to inhibit production of melanin in the mouse melanoma cell line B16F10. Importantly, unlike most glycosidase inhibitors, mangostenone F displayed very low cytotoxicity (EC50 > 200 μM). The Western blot for expression levels of proteins involved in melanogenesis showed that mangostenone F down-regulated tyrosinase and TRP-2 expression. Treating B16F10 cells with mangostenone F significantly increased the susceptibility of tyrosinase to endoglycosidase H digestion, indicating that tyrosinase was unable to mature fully and pass to the trans-golgi apparatus. Consistent with these data, in lysate assays, mangostenone F was shown to be a better inhibitor of α-glucosidases than deoxynojirimycin, a representative glycosidase inhibitor.
Collapse
Affiliation(s)
- Hyung Won Ryu
- Division of Applied Life Science (BK21 Program), IALS, Graduate School of Gyeongsang National University , Jinju 660-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
15
|
da Rocha DR, Santos WC, Lima ES, Ferreira VF. Synthesis of 1,2,3-triazole glycoconjugates as inhibitors of α-glucosidases. Carbohydr Res 2012; 350:14-9. [DOI: 10.1016/j.carres.2011.12.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/22/2011] [Accepted: 12/25/2011] [Indexed: 10/14/2022]
|
16
|
Stütz AE, Wrodnigg TM. Imino sugars and glycosyl hydrolases: historical context, current aspects, emerging trends. Adv Carbohydr Chem Biochem 2011; 66:187-298. [PMID: 22123190 DOI: 10.1016/b978-0-12-385518-3.00004-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Forty years of discoveries and research on imino sugars, which are carbohydrate analogues having a basic nitrogen atom instead of oxygen in the sugar ring and, acting as potent glycosidase inhibitors, have made considerable impact on our contemporary understanding of glycosidases. Imino sugars have helped to elucidate the catalytic machinery of glycosidases and have refined our methods and concepts of utilizing them. A number of new aspects have emerged for employing imino sugars as pharmaceutical compounds, based on their profound effects on metabolic activities in which glycosidases are involved. From the digestion of starch to the fight against viral infections, from research into malignant diseases to potential improvements in hereditary storage disorders, glycosidase action and inhibition are essential issues. This account aims at combining general developments with a focus on some niches where imino sugars have become useful tools for glycochemistry and glycobiology.
Collapse
Affiliation(s)
- Arnold E Stütz
- Institut für Organische Chemie, Technische Universität Graz, Austria
| | | |
Collapse
|