1
|
Agarwal U, Verma S, Tonk RK. Chromenone: An emerging scaffold in anti-Alzheimer drug discovery. Bioorg Med Chem Lett 2024; 111:129912. [PMID: 39089526 DOI: 10.1016/j.bmcl.2024.129912] [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: 05/05/2024] [Revised: 07/11/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Alzheimer's disease (AD) presents a growing global health concern. In recent decades, natural and synthetic chromenone have emerged as promising drug candidates due to their multi-target potential. Natural chromenone, quercetin, scopoletin, esculetin, coumestrol, umbelliferone, bergapten, and methoxsalen (xanthotoxin), and synthetic chromenone hybrids comprising structures like acridine, 4-aminophenyl, 3-arylcoumarins, quinoline, 1,3,4-oxadiazole, 1,2,3-triazole, and tacrine, have been explored for their potential to combat AD. Key reactions used for synthesis of chromenone hybrids include Perkin and Pechmann condensation. The activity of chromenone hybrids has been reported against several drug targets, including AChE, BuChE, BACE-1, and MAO-A/B. This review comprehensively explores natural, semisynthetic, and synthetic chromenone, elucidating their synthetic routes, possible mode of action/drug targets and structure-activity relationships (SAR). The acquired knowledge provides valuable insights for the development of new chromenone hybrids against AD.
Collapse
Affiliation(s)
- Uma Agarwal
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences & Research University, Delhi 110017, India
| | - Saroj Verma
- Pharmaceutical Chemistry Division, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram 122103, India.
| | - Rajiv K Tonk
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences & Research University, Delhi 110017, India.
| |
Collapse
|
2
|
Patyra A, Vaillé J, Omhmmed S, Dudek MK, Neasta J, Kiss AK, Oiry C. Pharmacological and phytochemical insights on the pancreatic β-cell modulation by Angelica L. roots. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118133. [PMID: 38580187 DOI: 10.1016/j.jep.2024.118133] [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: 02/09/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Angelica roots are a significant source of traditional medicines for various cultures around the northern hemisphere, from indigenous communities in North America to Japan. Among its many applications, the roots are used to treat type 2 diabetes mellitus; however, this application is not mentioned often. Ethnopharmacological studies have reported the use of A. japonica var. hirsutiflora, A. furcijuga, A. shikokiana, and A. keiskei to treat diabetes symptoms, and further reports have demonstrated the three angelica roots, i.e., A. japonica var. hirsutiflora, A. reflexa, and A. dahurica, exhibit insulin secretagogue activity. AIM OF THE STUDY This study aimed to phytochemically characterize and compare angelica roots monographed in the European Pharmacopeia 11th, isolate major plant metabolites, and assess extracts and isolates' capability to modulate pancreatic β-cell function. MATERIALS AND METHODS Root extracts of Angelica archangelica, Angelica dahurica, Angelica biserrata, and Angelica sinensis were phytochemically profiled using liquid chromatography method coupled with mass spectrometry. Based on this analysis, simple and furanocoumarins were isolated using chromatography techniques. Extracts (1.6-50 μg/mL) and isolated compounds (5-40 μmol/L) were studied for their ability to modulate insulin secretion in the rat insulinoma INS-1 pancreatic β-cell model. Insulin was quantified by the homogeneous time-resolved fluorescence method. RESULTS Forty-one secondary metabolites, mostly coumarins, were identified in angelica root extracts. A. archangelica, A. dahurica, and A. biserrata root extracts at concentration of 12.5-50 μg/mL potentiated glucose-induced insulin secretion, which correlated with their high coumarin content. Subsequently, 23 coumarins were isolated from these roots and screened using the same protocol. Coumarins substituted with the isoprenyl group were found to be responsible for the extracts' insulinotropic effect. CONCLUSIONS Insulinotropic effects of three pharmacopeial angelica roots were found, the metabolite profiles and pharmacological activities of the roots were correlated, and key structures responsible for the modulation of pancreatic β-cell function were identified. These findings may have implications for the traditional use of angelica roots in treating diabetes. Active plant metabolites may also become lead structures in the search for new antidiabetic treatments.
Collapse
Affiliation(s)
- Andrzej Patyra
- IBMM, Univ Montpellier, CNRS, ENSCM, 34293, Montpellier, France; Department of Pharmaceutical Biology, Medical University of Warsaw, 02-097, Warsaw, Poland.
| | - Justine Vaillé
- IBMM, Univ Montpellier, CNRS, ENSCM, 34293, Montpellier, France.
| | - Soufiyan Omhmmed
- IBMM, Univ Montpellier, CNRS, ENSCM, 34293, Montpellier, France.
| | - Marta Katarzyna Dudek
- Structural Studies Department, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-001, Łódź, Poland.
| | - Jérémie Neasta
- IBMM, Univ Montpellier, CNRS, ENSCM, 34293, Montpellier, France.
| | - Anna Karolina Kiss
- Department of Pharmaceutical Biology, Medical University of Warsaw, 02-097, Warsaw, Poland.
| | - Catherine Oiry
- IBMM, Univ Montpellier, CNRS, ENSCM, 34293, Montpellier, France.
| |
Collapse
|
3
|
Pan Y, Zhou M, Liu Z, Hao C, Zhai J, Liu R, Shi Z, Sun J, Wang X. Synthesis and activity of arylcoumarin derivatives with therapeutic effects on diabetic nephropathy. Arch Pharm (Weinheim) 2024; 357:e2300524. [PMID: 38036297 DOI: 10.1002/ardp.202300524] [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: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023]
Abstract
In the literature, daidzein has been reported to exhibit cardiovascular protective effects and hypoglycemic activity in mice. We sought to design and synthesize a novel compound, SJ-6, an analog of daidzein, with improved hypoglycemic properties. Although SJ-6 demonstrated favorable hypoglycemic effects, its pharmacokinetic limitations prompted us to design and synthesize prodrugs of SJ-6. We conducted a comprehensive evaluation of the prodrugs, including in vitro and in vivo studies, such as cytotoxicity, absorption, distribution, metabolism, excretion, and toxicity (ADMET) simulation analysis, in vitro blood-brain barrier (BBB) permeability evaluation, compound effect on insulin resistance, oral glucose tolerance test (OGTT), in vivo plasma concentration testing, acute toxicity test in rats, and long-term gavage administration experiment. Furthermore, we examined the antidiabetic nephropathy activity of our lead compound, compound 10, which demonstrated superior efficacy compared with the positive control drug, metformin hydrochloride. Our findings suggest that compound 10 represents a promising lead compound for the prevention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Yinbo Pan
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
- School of Chemistry and Chemical Engineering, School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Min Zhou
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Zhenzhen Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Canhua Hao
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Jingfang Zhai
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Ren Liu
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Zezhou Shi
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Jie Sun
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Xiaojing Wang
- School of Parmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| |
Collapse
|
4
|
Tariq HZ, Saeed A, Ullah S, Fatima N, Halim SA, Khan A, El-Seedi HR, Ashraf MZ, Latif M, Al-Harrasi A. Synthesis of novel coumarin-hydrazone hybrids as α-glucosidase inhibitors and their molecular docking studies. RSC Adv 2023; 13:26229-26238. [PMID: 37670997 PMCID: PMC10475976 DOI: 10.1039/d3ra03953f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/17/2023] [Indexed: 09/07/2023] Open
Abstract
Diabetes mellitus is a metabolic disorder and more than 90% of diabetic patients suffer from type-2 diabetes, which is characterized by hyperglycemia. α-Glucosidase inhibition has become an appropriate approach to tackle high blood glucose levels. The current study was focused on synthesizing coumarin-hydrazone hybrids (7a-i) by using facile chemical reactions. The synthesized compounds were characterized by using 1H-NMR, 13C-NMR, and IR. To evaluate their anti-diabetic capability, all of the conjugates were screened for in vitro α-glucosidase inhibitory activity to reveal their therapeutic importance. All of the compounds (except 7b) demonstrated significant enzyme inhibitory potential with IC50 values ranging between 2.39-57.52 μM, as compared to the standard inhibitor, acarbose (IC50 = 873.34 ± 1.67 μM). Among them, compound 7c is the most potent α-glucosidase inhibitor (IC50 = 2.39 ± 0.05 μM). Additionally, molecular docking was employed to scrutinize the binding pattern of active compounds within the α-glucosidase binding site. The in silico analysis reflects that hydrazone moiety is an essential pharmacophore for the binding of compounds with the active site residues of the enzyme. This study demonstrates that compounds 7c and 7f deserve further molecular optimization for potential application in diabetic management.
Collapse
Affiliation(s)
- Hafiza Zara Tariq
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-51-9064-2128
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-51-9064-2128
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Noor Fatima
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-51-9064-2128
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Hesham R El-Seedi
- School of Food and Biological Engineering, Jiangsu University Zhenjiang 212013 China
- Department of Chemistry, Faculty of Science, Menoufia University Shebin El-Kom 32512 Egypt
| | | | - Muhammad Latif
- Centre for Genetics and Inherited Diseases (CGID), Taibah University Al-Madinah Al-Munawwarah Kingdom of Saudi Arabia
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| |
Collapse
|
5
|
Shafiq N, Shahzad N, Rida F, Ahmad Z, Nazir HA, Arshad U, Zareen G, Attiq N, Parveen S, Rashid M, Ali B. One-pot multicomponent synthesis of novel pyridine derivatives for antidiabetic and antiproliferative activities. Future Med Chem 2023; 15:1069-1089. [PMID: 37503685 DOI: 10.4155/fmc-2023-0132] [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] [Indexed: 07/29/2023] Open
Abstract
Background: Due to the close relationship of diabetes with hypertension reported in various research, a set of pyridine derivatives with US FDA-approved drug cores were designed and integrated by artificial intelligence. Methods: Novel pyridines were designed and synthesized. Compounds MNS-1-MNS-4 were evaluated for their structure and were screened for their in vitro antidiabetic (α-amylase) activity and anticancer (HepG2) activity by methyl thiazolyl tetrazolium assay. Comparative 3D quantitative structure-activity relationship analysis and pharmacophore generation were carried out. Results: The study revealed MNS-1 and MNS-4 as good alternatives to acarbose as antidiabetic agents, and MNS-2 as a more viable, better alternative to doxorubicin in the methyl thiazolyl tetrazolium assay. Conclusion: This combination of studies identifies new and more active analogs of existing FDA-approved drugs for the treatment of diabetes.
Collapse
Affiliation(s)
- Nusrat Shafiq
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Nabeel Shahzad
- Department of Chemistry, University of WAH, Wah Cantt, 44700, Pakistan
| | - Fatima Rida
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Zaheer Ahmad
- Department of Chemistry, University of WAH, Wah Cantt, 44700, Pakistan
| | - Hafiza Ayesha Nazir
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Uzma Arshad
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Gul Zareen
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Naila Attiq
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Shagufta Parveen
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Maryam Rashid
- Synthetic & Natural Product Discovery Laboratory, Department of Chemistry, Government College Women's University Faisalabad, 38000, Pakistan
| | - Basharat Ali
- Department of Chemistry, Khawaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, Punjab, 64200, Pakistan
| |
Collapse
|
6
|
Mahnashi MH, Alam W, Huneif MA, Abdulwahab A, Alzahrani MJ, Alshaibari KS, Rashid U, Sadiq A, Jan MS. Exploration of Succinimide Derivative as a Multi-Target, Anti-Diabetic Agent: In Vitro and In Vivo Approaches. Molecules 2023; 28:molecules28041589. [PMID: 36838577 PMCID: PMC9964140 DOI: 10.3390/molecules28041589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Diabetes mellitus (DM) is counted among one of the leading challenges in the recent era, and it is a life-threatening disorder. Compound 4-hydroxy 3-methoxy phenylacetone (compound 1) was previously isolated from Polygonum aviculare. This compound was reacted with N-benzylmaleimide to synthesize the targeted compound 3. The purpose of this research is to exhibit our developed compound 3's ability to concurrently inhibit many targets that are responsible for hyperglycemia. Compound 3 was capable of inhibiting α-amylase, α-glucosidase, and protein tyrosine phosphatase 1 B. Even so, outstanding in vitro inhibition was shown by the compound against dipeptidyl peptidase-4 (DPP-4) with an IC50 value of 0.07 µM. Additionally, by using DPPH in the antioxidant activity, it exhibited good antioxidant potential. Similarly, in the in vivo activity, the experimental mice proved to be safe by treatment with compound 3. After 21 days of examination, the compound 3 activity pattern was found to be effective in experimental mice. Compound 3 decreased the excess peak of total triglycerides, total cholesterol, AST, ALT, ALP, LDL, BUN, and creatinine in the STZ-induced diabetic mice. Likewise, the histopathology of the kidneys, liver, and pancreas of the treated animals was also evaluated. Overall, the succinimde moiety, such as compound 3, can affect several targets simultaneously, and, finally, we were successful in synthesizing a multi-targeted preclinical therapy.
Collapse
Affiliation(s)
- Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran 55461, Saudi Arabia
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Mohammed A. Huneif
- Pediatric Department, Medical College, Najran University, Najran 55461, Saudi Arabia
| | - Alqahtani Abdulwahab
- Pediatric Department, Medical College, Najran University, Najran 55461, Saudi Arabia
| | | | - Khaled S. Alshaibari
- Pediatric Department, Medical College, Najran University, Najran 55461, Saudi Arabia
| | - Umar Rashid
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Chakdara 18800, Pakistan
- Correspondence: (A.S.); (M.S.J.)
| | - Muhammad Saeed Jan
- Department of Pharmacy, Bacha Khan University, Charsadda 24420, Pakistan
- Correspondence: (A.S.); (M.S.J.)
| |
Collapse
|
7
|
Recent developments in synthetic α-glucosidase inhibitors: A comprehensive review with structural and molecular insight. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
8
|
Li Y, Pan Y, Wang L, Wang X, Chu H, Li Y, Mu Y, Sun J. 3-Arylcoumarin inhibits vascular calcification by inhibiting the generation of AGEs and anti-oxidative stress. J Enzyme Inhib Med Chem 2022; 37:2147-2157. [PMID: 35950567 PMCID: PMC9377248 DOI: 10.1080/14756366.2022.2109024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective This work aims to screen drugs for preventing and treating vascular calcification. Method: We screened a series of 3-arylcoumarins for the detection of vascular calcification-associated factors using human aortic vascular smooth muscle cells. Results We found that compounds 14 and 32 significantly inhibited alkaline phosphatase (ALP) activity similar to aminoguanidine hydrochloride (AGH) in a cellular model of AGEs-induced calcification. We also found that compounds 14 and 32 could significantly decrease the levels of factors such as AGEs, intracellular calcium ions, and total ROS in the calcified cell model. Further study indicates that compound 14 could significantly inhibit the expression of P-ERK1/2, PKC, NF-κB, RAGE and OPN proteins and increased the expression of SM22-α and PPAR-γ proteins in the calcified cells. Conclusion We speculate that compound 14 inhibits vascular calcification by inhibiting oxidative stress and inhibiting AGEs production, suggesting that 3-arylcoumarin derivatives are potential candidates for the treatment of vascular calcification. Vascular calcification is a process similar to bone formation, which is highly adjustable and active. Currently, there are no specific drugs to delay or reverse vascular calcification. Through the screening of 44 coumarin compounds synthesised by our group, compound 14 was obtained to dose-dependently inhibit the calcification of vascular smooth muscle cells without affecting the normal proliferation of cells, decreasing the intracellular calcium concentration, inhibiting the activity of ALP enzyme. In conclusion, the calcium lowering effect of compound 14 is a potential candidate for drugs for the treatment of vascular calcification.
Collapse
Affiliation(s)
- YuFei Li
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yinbo Pan
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Liying Wang
- Shandong Electric Power Central Hospital, Jinan, China
| | - Xiaojing Wang
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haiping Chu
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yan Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yanling Mu
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Sun
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
9
|
Tsivileva OM, Koftin OV, Evseeva NV. Coumarins as Fungal Metabolites with Potential Medicinal Properties. Antibiotics (Basel) 2022; 11:1156. [PMID: 36139936 PMCID: PMC9495007 DOI: 10.3390/antibiotics11091156] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Coumarins are a structurally varied set of 2H-chromen-2-one compounds categorized also as members of the benzopyrone group of secondary metabolites. Coumarin derivatives attract interest owing to their wide practical application and the unique reactivity of fused benzene and pyrone ring systems in molecular structure. Coumarins have their own specific fingerprints as antiviral, antimicrobial, antioxidant, anti-inflammatory, antiadipogenic, cytotoxic, apoptosis, antitumor, antitubercular, and cytotoxicity agents. Natural products have played an essential role in filling the pharmaceutical pipeline for thousands of years. Biological effects of natural coumarins have laid the basis of low-toxic and highly effective drugs. Presently, more than 1300 coumarins have been identified in plants, bacteria, and fungi. Fungi as cultivated microbes have provided many of the nature-inspired syntheses of chemically diverse drugs. Endophytic fungi bioactivities attract interest, with applications in fields as diverse as cancer and neuronal injury or degeneration, microbial and parasitic infections, and others. Fungal mycelia produce several classes of bioactive molecules, including a wide group of coumarins. Of promise are further studies of conditions and products of the natural and synthetic coumarins' biotransformation by the fungal cultures, aimed at solving the urgent problem of searching for materials for biomedical engineering. The present review evaluates the fungal coumarins, their structure-related peculiarities, and their future therapeutic potential. Special emphasis has been placed on the coumarins successfully bioprospected from fungi, whereas an industry demand for the same coumarins earlier found in plants has faced hurdles. Considerable attention has also been paid to some aspects of the molecular mechanisms underlying the coumarins' biological activity. The compounds are selected and grouped according to their cytotoxic, anticancer, antibacterial, antifungal, and miscellaneous effects.
Collapse
Affiliation(s)
- Olga M. Tsivileva
- Laboratory of Microbiology, Institute of Biochemistry and Physiology of Plants and Microorganisms, Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Oleg V. Koftin
- Department of Biochemistry, V.I. Razumovsky Saratov State Medical University, 112 ul. Bol’shaya Kazach’ya, Saratov 410012, Russia
| | - Nina V. Evseeva
- Laboratory of Immunochemistry, Institute of Biochemistry and Physiology of Plants and Microorganisms, Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospekt Entuziastov, Saratov 410049, Russia
| |
Collapse
|
10
|
Zhang Q, Miao YH, Liu T, Yun YL, Sun XY, Yang T, Sun J. Natural source, bioactivity and synthesis of 3-Arylcoumarin derivatives. J Enzyme Inhib Med Chem 2022; 37:1023-1042. [PMID: 35438580 PMCID: PMC9037183 DOI: 10.1080/14756366.2022.2058499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
3-arylcoumarins with different pharmacological properties widely exist in a variety of natural plants. The extensive research on 3-arylcoumarins was attributed to its therapeutic and relatively easy isolation. Therefore, 3-arylcoumarins can be recognised as useful structures for the design of novel compounds with potential pharmacological interest, particularly in the fields of anti-inflammatory, anti-cancer, antioxidant, Monoamine oxidase (MAO) enzyme inhibition, etc. The current review highlights the biological activities, design, and chemical synthetic methods of 3-arylcoumarins derivatives as well as their important natural product sources.
Collapse
Affiliation(s)
- Qiang Zhang
- Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yu-Hang Miao
- Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Teng Liu
- Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yin-Ling Yun
- Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao-Ya Sun
- Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tao Yang
- Department of Thoracic and Cardiovascular Surgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - Jie Sun
- Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
11
|
Rossi R, Ciofalo M. Palladium-Catalysed Intermolecular Direct C–H Bond Arylation of Heteroarenes with Reagents Alternative to Aryl Halides: Current State of the Art. CURR ORG CHEM 2022. [DOI: 10.2174/1385272826666220201124008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Abstract: This unprecedented review with 322 references provides a critical up-to-date picture of the Pd-catalysed intermolecular direct C–H bond arylation of heteroarenes with arylating reagents alternative to aryl halides that include aryl sulfonates (aryl triflates, tosylates, mesylates, and imidazole-1-sulfonates), diaryliodonium salts, [(diacetoxy)iodo]arenes, arenediazonium salts, 1-aryltriazenes, arylhydrazines and N’-arylhydrazides, arenesulfonyl chlorides, sodium arenesulfinates, arenesulfinic acids, and arenesulfonohydrazides. Particular attention has been paid to summarise the preparation of the various arylating reagents and to highlight the practicality, versatility, and limitations of the various developed arylation protocols, also comparing their results with those achieved in analogous Pd-catalysed arylation reactions involving the use of aryl halides as electrophiles. Mechanistic proposals have also been briefly summarised and discussed. However, data concerning Pd-catalysed direct C–H bond arylations involving the C–H bonds of aryl substituents of the examined heteroarene derivatives have not been taken into account.
Collapse
Affiliation(s)
- Renzo Rossi
- Dipartimento di Chimica e Chimica Industriale, University of Pisa, Via G. Moruzzi 3, I-56124 Pisa, Italy
| | - Maurizio Ciofalo
- Dipartimento di Scienze Agrarie, Alimentari e Forestali, University of Palermo, Viale delle Scienze, Edificio 4, I-90128, Palermo, Italy
| |
Collapse
|
12
|
Pan Y, Liu T, Wang X, Sun J. Research progress of coumarins and their derivatives in the treatment of diabetes. J Enzyme Inhib Med Chem 2022; 37:616-628. [PMID: 35067136 PMCID: PMC8788346 DOI: 10.1080/14756366.2021.2024526] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Diabetes is a group of metabolic diseases characterised by chronic hyperglycaemia caused by multiple causes, which is caused by insulin secretion and/or utilisation defects. It is characterised by increased fasting and postprandial blood glucose levels due to insulin deficiency or insulin resistance. It is reported that the harm of diabetes mainly comes from its complications, and the cardiovascular disease caused by diabetes is the primary cause of its harm. China has the largest number of diabetic patients in the world, and the prevention and control of diabetes are facing great challenges. In recent years, many kinds of literature have been published abroad, which have proved that coumarin and its derivatives are effective in the treatment of diabetic complications such as nephropathy and cardiovascular disease. In this paper, the types of antidiabetic drugs and the anti-diabetic mechanism of coumarins were reviewed.
Collapse
Affiliation(s)
- Yinbo Pan
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University, Jinan, Shandong, China
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Teng Liu
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University, Jinan, Shandong, China
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University, Jinan, Shandong, China
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Sun
- School of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University, Jinan, Shandong, China
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
13
|
Matos MJ, Uriarte E, Santana L. 3-Phenylcoumarins as a Privileged Scaffold in Medicinal Chemistry: The Landmarks of the Past Decade. Molecules 2021; 26:6755. [PMID: 34771164 PMCID: PMC8587835 DOI: 10.3390/molecules26216755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/27/2022] Open
Abstract
3-Phenylcoumarins are a family of heterocyclic molecules that are widely used in both organic and medicinal chemistry. In this overview, research on this scaffold, since 2010, is included and discussed, focusing on aspects related to its natural origin, synthetic procedures and pharmacological applications. This review paper is based on the most relevant literature related to the role of 3-phenylcoumarins in the design of new drug candidates. The references presented in this review have been collected from multiple electronic databases, including SciFinder, Pubmed and Mendeley.
Collapse
Affiliation(s)
- Maria J Matos
- Centro de Investigação em Química da Universidade do Porto (CIQUP), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eugenio Uriarte
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Instituto de Ciencias Químicas Aplicadas, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Lourdes Santana
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
14
|
Zhang YX, Ao Z, He YW, Lu JY, Chen XL, Kong LY, Luo JG. Hyperpatulones C-G, new spirocyclic polycyclic polyprenylated acylphloroglucinols from the leaves of Hypericum patulum. Fitoterapia 2021; 155:105063. [PMID: 34655700 DOI: 10.1016/j.fitote.2021.105063] [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: 09/05/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 11/25/2022]
Abstract
Five new spirocyclic polycyclic polyprenylated acylphloroglucinols, Hyperpatulones C-G (1-5), were obtained from the leaves of Hypericum patulum. Their structures were characterized by the comprehensive analysis of their IR, NMR, CD spectra and HRESIMS data. All the new compounds were evaluated for the α-glycosidase inhibitory activities. Among them, compounds 3-5 showed α-glucosidase inhibitory activities, with IC50 values of 14.06-37.69 μM.
Collapse
Affiliation(s)
- Yu-Xin Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Zhen Ao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Yi-Wen He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Jin-Yu Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Xin-Lin Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Ling-Yi Kong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| | - Jian-Guang Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| |
Collapse
|
15
|
Coumarins as Tool Compounds to Aid the Discovery of Selective Function Modulators of Steroid Hormone Binding Proteins. MOLECULES (BASEL, SWITZERLAND) 2021; 26:molecules26175142. [PMID: 34500576 PMCID: PMC8433903 DOI: 10.3390/molecules26175142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 11/20/2022]
Abstract
Steroid hormones play an essential role in a wide variety of actions in the body, such as in metabolism, inflammation, initiating and maintaining sexual differentiation and reproduction, immune functions, and stress response. Androgen, aromatase, and sulfatase pathway enzymes and nuclear receptors are responsible for steroid biosynthesis and sensing steroid hormones. Changes in steroid homeostasis are associated with many endocrine diseases. Thus, the discovery and development of novel drug candidates require a detailed understanding of the small molecule structure–activity relationship with enzymes and receptors participating in steroid hormone synthesis, signaling, and metabolism. Here, we show that simple coumarin derivatives can be employed to build cost-efficiently a set of molecules that derive essential features that enable easy discovery of selective and high-affinity molecules to target proteins. In addition, these compounds are also potent tool molecules to study the metabolism of any small molecule.
Collapse
|
16
|
Peytam F, Takalloobanafshi G, Saadattalab T, Norouzbahari M, Emamgholipour Z, Moghimi S, Firoozpour L, Bijanzadeh HR, Faramarzi MA, Mojtabavi S, Rashidi-Ranjbar P, Karima S, Pakraad R, Foroumadi A. Design, synthesis, molecular docking, and in vitro α-glucosidase inhibitory activities of novel 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines against yeast and rat α-glucosidase. Sci Rep 2021; 11:11911. [PMID: 34099819 PMCID: PMC8184976 DOI: 10.1038/s41598-021-91473-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
In an attempt to find novel, potent α-glucosidase inhibitors, a library of poly-substituted 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines 3a-ag have been synthesized through heating a mixture of 2-aminobenzimidazoles 1 and α-azidochalcone 2 under the mild conditions. This efficient, facile protocol has been resulted into the desirable compounds with a wide substrate scope in good to excellent yields. Afterwards, their inhibitory activities against yeast α-glucosidase enzyme were investigated. Showing IC50 values ranging from 16.4 ± 0.36 µM to 297.0 ± 1.2 µM confirmed their excellent potency to inhibit α-glucosidase which encouraged us to perform further studies on α-glucosidase enzymes obtained from rat as a mammal source. Among various synthesized 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines, compound 3k exhibited the highest potency against both Saccharomyces cerevisiae α-glucosidase (IC50 = 16.4 ± 0.36 μM) and rat small intestine α-glucosidase (IC50 = 45.0 ± 8.2 μM). Moreover, the role of amine moiety on the observed activity was studied through substituting with chlorine and hydrogen resulted into a considerable deterioration on the inhibitory activity. Kinetic study and molecular docking study have confirmed the in-vitro results.
Collapse
Affiliation(s)
- Fariba Peytam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Toktam Saadattalab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Norouzbahari
- Faculty of Medicine, Eastern Mediterranean University, via Mersin 10, Famagusta, Northern Cyprus, Turkey
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Bijanzadeh
- Department of Environmental Sciences, Faculty of Natural Resources and Marine Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, 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
| | | | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Roya Pakraad
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Maurya AK, Mulpuru V, Mishra N. Discovery of Novel Coumarin Analogs against the α-Glucosidase Protein Target of Diabetes Mellitus: Pharmacophore-Based QSAR, Docking, and Molecular Dynamics Simulation Studies. ACS OMEGA 2020; 5:32234-32249. [PMID: 33376861 PMCID: PMC7758891 DOI: 10.1021/acsomega.0c03871] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/10/2020] [Indexed: 05/13/2023]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease, the third killer of mankind. The finding of potent drugs against diabetes remains challenging. In the present study, coumarin derivatives with known biological activity against diabetic protein have been used to predict functional groups' positions on coumarin derivatives. α-Glucosidase is a brush border membrane-bound lysosomal enzyme from the hydrolase enzyme family. It plays an important role in the metabolism of glycoproteins. Inhibitors of lysosomal α-glucosidase can reduce postprandial hyperglycemia. Due to this, lysosomal α-glucosidase is a good therapeutic target for drugs. A total of 116 coumarin derivatives with IC50 values against lysosomal α-glucosidase were selected for a CADD (computer-aided drug design) approach to identify more potent drugs. Pharmacophore modeling and atom-based 3-QSAR of 116 active compounds against lysosomal α-glucosidase were performed and identified positions and types of groups to increase activity. We performed molecular docking of 116 coumarin derivatives against the lysosomal α-glucosidase enzyme, and three compounds (isorutarine, 10_, and 36) resulted in a docking score of -7.64, -7.12, and -6.86 kcal/mol. The molecular dynamics simulation of the above three molecules and protein complex performed for 100 ns supported the interaction stability of isorutarine, 10_, and 36 with the lysosomal binding site α-glucosidase.
Collapse
|
18
|
Tafesse TB, Bule MH, Khoobi M, Faramarzi MA, Abdollahi M, Amini M. Coumarin-based Scaffold as α-glucosidase Inhibitory Activity: Implication for the Development of Potent Antidiabetic Agents. Mini Rev Med Chem 2020; 20:134-151. [PMID: 31553294 DOI: 10.2174/1389557519666190925162536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/15/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Delaying the absorption of glucose through α-glucosidase enzyme inhibition is one of the therapeutic approaches in the management of Type 2 diabetes, which can reduce the incidence of postprandial hyperglycemia. The existence of chronic postprandial hyperglycemia impaired the endogenous antioxidant defense by inducing oxidative stress-induced pancreatic β-cell destruction through uncontrolled generation of free radicals such as ROS, which in turn, leads to various macrovascular and microvascular complications. The currently available α -glucosidase inhibitors, for instance, acarbose, have some side effects such as hypoglycemia at higher doses, liver problems, meteorism, diarrhea, and lactic acidosis. Therefore, there is an urgent need to discover and develop potential α-glucosidase inhibitors. OBJECTIVE Based on suchmotifs, researchers are intrigued to search for the best scaffold that displays various biological activities. Among them, coumarin scaffold has attracted great attention. The compound and its derivatives can be isolated from various natural products and/or synthesized for the development of novel α-glucosidase inhibitors. RESULTS This study focused on coumarin and its derivatives as well as on their application as potent antidiabetic agents and has also concentrated on the structure-activity relationship. CONCLUSION This review describes the applications of coumarin-containing derivatives as α - glucosidase inhibitors based on published reports which will be useful for innovative approaches in the search for novel coumarin-based antidiabetic drugs with less toxicity and more potency.
Collapse
Affiliation(s)
- Tadesse Bekele Tafesse
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences-International Campus (IC-TUMS), Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,School of Pharmacy, College of Health & Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mohammed Hussen Bule
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences-International Campus (IC-TUMS), Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacy, College of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia
| | - Mehdi Khoobi
- Department of Pharmaceutical Biomaterials, Medical Biomaterials Research Center and The Institute of Pharmaceutical Sciences (TIPS), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center and The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Annunziata F, Pinna C, Dallavalle S, Tamborini L, Pinto A. An Overview of Coumarin as a Versatile and Readily Accessible Scaffold with Broad-Ranging Biological Activities. Int J Mol Sci 2020; 21:E4618. [PMID: 32610556 PMCID: PMC7370201 DOI: 10.3390/ijms21134618] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/22/2020] [Accepted: 06/28/2020] [Indexed: 12/19/2022] Open
Abstract
Privileged structures have been widely used as an effective template for the research and discovery of high value chemicals. Coumarin is a simple scaffold widespread in Nature and it can be found in a considerable number of plants as well as in some fungi and bacteria. In the last years, these natural compounds have been gaining an increasing attention from the scientific community for their wide range of biological activities, mainly due to their ability to interact with diverse enzymes and receptors in living organisms. In addition, coumarin nucleus has proved to be easily synthetized and decorated, giving the possibility of designing new coumarin-based compounds and investigating their potential in the treatment of various diseases. The versatility of coumarin scaffold finds applications not only in medicinal chemistry but also in the agrochemical field as well as in the cosmetic and fragrances industry. This review is intended to be a critical overview on coumarins, comprehensive of natural sources, metabolites, biological evaluations and synthetic approaches.
Collapse
Affiliation(s)
- Francesca Annunziata
- Department of Pharmaceutical Science, University of Milan, via Mangiagalli 25, 20133 Milan, Italy; (F.A.); (C.P.)
| | - Cecilia Pinna
- Department of Pharmaceutical Science, University of Milan, via Mangiagalli 25, 20133 Milan, Italy; (F.A.); (C.P.)
| | - Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences, University of Milan, via Celoria 2, 20133 Milan, Italy; (S.D.); (A.P.)
| | - Lucia Tamborini
- Department of Pharmaceutical Science, University of Milan, via Mangiagalli 25, 20133 Milan, Italy; (F.A.); (C.P.)
| | - Andrea Pinto
- Department of Food, Environmental and Nutritional Sciences, University of Milan, via Celoria 2, 20133 Milan, Italy; (S.D.); (A.P.)
| |
Collapse
|
20
|
Gupta MK, Kumar S, Chaudhary S. Synthesis and Investigation of Antidiabetic Response of New Coumarin Derivatives Against Streptozotocin Induced Diabetes in Experimental Rats. Pharm Chem J 2020. [DOI: 10.1007/s11094-020-02134-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
21
|
Dinparast L, Hemmati S, Zengin G, Alizadeh AA, Bahadori MB, Kafil HS, Dastmalchi S. Rapid, Efficient, and Green Synthesis of Coumarin Derivatives via Knoevenagel Condensation and Investigating Their Biological Effects. ChemistrySelect 2019. [DOI: 10.1002/slct.201901921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Leila Dinparast
- Biotechnology Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Salar Hemmati
- Drug Applied Research CenterTabriz University of Medical Sciences, Tabriz Iran
| | - Gokhan Zengin
- Department of BiologyScience FacultySelcuk University Konya Turkey
| | - Ali Akbar Alizadeh
- Biotechnology Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Mir Babak Bahadori
- Medicinal Plants Research CenterMaragheh University of Medical Sciences Maragheh Iran
| | | | - Siavoush Dastmalchi
- Biotechnology Research CenterTabriz University of Medical Sciences Tabriz Iran
- School of PharmacyTabriz University of Medical Sciences Tabriz Iran
- Faculty of PharmacyNear East University, POBOX:99138 Nicosia, North Cyprus, Mersin 10 Turkey
| |
Collapse
|
22
|
Dan WJ, Zhang Q, Zhang F, Wang WW, Gao JM. Benzonate derivatives of acetophenone as potent α-glucosidase inhibitors: synthesis, structure-activity relationship and mechanism. J Enzyme Inhib Med Chem 2019; 34:937-945. [PMID: 31072245 PMCID: PMC6522914 DOI: 10.1080/14756366.2019.1604519] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In this article, 23 compounds (6 and 7a–7v) were prepared and evaluated for their in vitro α-glucosidase inhibitory activity. The compounds 7d, 7f, 7i, 7n, 7o, 7r, 7s, 7u, and 7v displayed the α-glucosidase inhibition activity with IC50 values ranging from 1.68 to 7.88 µM. Among all tested compounds, 7u was found to be the most efficient, being 32-fold more active than the standard drug acarbose, which significantly attenuated postprandial blood glucose in mice. In addition, the compound 7u also induced the fluorescence quenching and conformational changes of enzyme, by forming α-glucosidase–7u complex in a mixed inhibition type. The thermodynamic constants recognised the interaction between 7u and α-glucosidase and was an enthalpy-driven spontaneous exothermic reaction. The synchronous fluorescence and CD spectra also indicate that the compound 7u changed the enzyme conformation. The findings identify the binding interactions between new ligands and α-glucosidase and reveal the compound 7u as a potent α-glucosidase inhibitor.
Collapse
Affiliation(s)
- Wen-Jia Dan
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Qiang Zhang
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Fan Zhang
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Wei-Wei Wang
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Jin-Ming Gao
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| |
Collapse
|
23
|
Saadat S, Beheshti F, Askari VR, Hosseini M, Mohamadian Roshan N, Boskabady MH. Aminoguanidine affects systemic and lung inflammation induced by lipopolysaccharide in rats. Respir Res 2019; 20:96. [PMID: 31113409 PMCID: PMC6530199 DOI: 10.1186/s12931-019-1054-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/22/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Nitric oxide is a mediator of potential importance in numerous physiological and inflammatory processes in the lung. Aminoguanidine (AG) has been shown to have anti-inflammation and radical scavenging properties. This study aimed to investigate the effects of AG, an iNOS inhibitor, on lipopolysaccharide (LPS)-induced systemic and lung inflammation in rats. METHODS Male Wistar rats were divided into control, LPS (1 mg/kg/day i.p.), and LPS groups treated with AG 50, 100 or 150 mg/kg/day i.p. for five weeks. Total nitrite concentration, total and differential white blood cells (WBC) count, oxidative stress markers, and the levels of IL-4, IFN-γ, TGF-β1, and PGE2 were assessed in the serum or bronchoalveolar lavage fluid (BALF). RESULTS Administration of LPS decreased IL-4 level (p < 0.01) in BALF, total thiol content, superoxide dismutase (SOD) and catalase (CAT) activities (p < 0.001) in BALF and serum, and increased total nitrite, malondialdehyde (MDA), IFN-γ, TGF-β1 and PGE2 (p < 0.001) concentrations in BALF. Pre-treatment with AG increased BALF level of IL-4 and total thiol as well as SOD and CAT activities (p < 0.05 to p < 0.001), but decreased BALF levels of total nitrite, MDA, IFN-γ, TGF-β1, and PGE2 (p < 0.01 to p < 0.001). AG treatment decreased total WBC count, lymphocytes and macrophages in BALF (p < 0.01 to p < 0.001) and improved lung pathological changes including interstitial inflammation and lymphoid infiltration (p < 0.05 to p < 0.001). CONCLUSIONS AG treatment reduced oxidant markers, inflammatory cytokines and lung pathological changes but increased antioxidants and anti-inflammatory cytokines. Therefore, AG may play a significant protective role against inflammation and oxidative stress that cause lung injury.
Collapse
Affiliation(s)
- Saeideh Saadat
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Vahid Reza Askari
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
- Student Research Committee, Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
| | - Nema Mohamadian Roshan
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hossein Boskabady
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.
| |
Collapse
|
24
|
Yang J, Zhang P, Hu Y, Liu T, Sun J, Wang X. Synthesis and biological evaluation of 3-arylcoumarins as potential anti-Alzheimer's disease agents. J Enzyme Inhib Med Chem 2019; 34:651-656. [PMID: 30746966 PMCID: PMC6374920 DOI: 10.1080/14756366.2019.1574297] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease, a neurodegenerative illness, has the extremely complex pathogenesis. Accumulating evidence indicates there is a close relationship between several enzymes and Alzheimer's disease. Various substituted 3-arylcoumarin derivatives were synthesised, and their in vitro activity, including cholinesterase inhibitory activity, monoamine oxidase inhibitory activity, and antioxidant activity were investigated. Most of the compounds exhibited high activity; therefore 3-arylcoumarin compounds have the potential as drug candidates for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jie Yang
- a School of Medicine and Life Sciences , University of Jinan-Shandong Academy of Medical Sciences , Jinan , China.,b Institute of MateriaMedica , Shandong Academy of Medical Sciences , Jinan , China.,c Key Laboratory for Biotech-Drugs Ministry of Health , Jinan , China.,d Key Laboratory for Rare & Uncommon Diseases of Shandong Province , Jinan , China
| | - Pingping Zhang
- a School of Medicine and Life Sciences , University of Jinan-Shandong Academy of Medical Sciences , Jinan , China.,b Institute of MateriaMedica , Shandong Academy of Medical Sciences , Jinan , China.,c Key Laboratory for Biotech-Drugs Ministry of Health , Jinan , China.,d Key Laboratory for Rare & Uncommon Diseases of Shandong Province , Jinan , China
| | - Yuheng Hu
- a School of Medicine and Life Sciences , University of Jinan-Shandong Academy of Medical Sciences , Jinan , China.,b Institute of MateriaMedica , Shandong Academy of Medical Sciences , Jinan , China.,c Key Laboratory for Biotech-Drugs Ministry of Health , Jinan , China.,d Key Laboratory for Rare & Uncommon Diseases of Shandong Province , Jinan , China
| | - Teng Liu
- a School of Medicine and Life Sciences , University of Jinan-Shandong Academy of Medical Sciences , Jinan , China.,b Institute of MateriaMedica , Shandong Academy of Medical Sciences , Jinan , China.,c Key Laboratory for Biotech-Drugs Ministry of Health , Jinan , China.,d Key Laboratory for Rare & Uncommon Diseases of Shandong Province , Jinan , China
| | - Jie Sun
- a School of Medicine and Life Sciences , University of Jinan-Shandong Academy of Medical Sciences , Jinan , China.,b Institute of MateriaMedica , Shandong Academy of Medical Sciences , Jinan , China.,c Key Laboratory for Biotech-Drugs Ministry of Health , Jinan , China.,d Key Laboratory for Rare & Uncommon Diseases of Shandong Province , Jinan , China
| | - Xiaojing Wang
- a School of Medicine and Life Sciences , University of Jinan-Shandong Academy of Medical Sciences , Jinan , China.,b Institute of MateriaMedica , Shandong Academy of Medical Sciences , Jinan , China.,c Key Laboratory for Biotech-Drugs Ministry of Health , Jinan , China.,d Key Laboratory for Rare & Uncommon Diseases of Shandong Province , Jinan , China
| |
Collapse
|