1
|
Giuliani C, Di Dalmazi G, Bucci I, Napolitano G. Quercetin and Thyroid. Antioxidants (Basel) 2024; 13:1202. [PMID: 39456456 PMCID: PMC11505551 DOI: 10.3390/antiox13101202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Quercetin is the most abundant flavonoid present in fruits and vegetables. For its antiproliferative, antiviral, anti-inflammatory and antioxidants activities, it is an active ingredient of several herbal remedies and is available as a nutraceutical. Experimental studies performed in vitro have demonstrated that quercetin inhibits growth and function in normal thyroid cells and may act as a thyroid disruptor. These effects have also been confirmed in vivo using rodent models. Some studies have reported the ability of quercetin to interfere with the metabolism of thyroid hormones, since it inhibits the 5'-deiodinase type 1 (D1) activity in the thyroid, as well as in the liver. Besides the effects on normal thyroid cells, several experiments performed in vitro have shown a potential therapeutic role of quercetin in thyroid cancer. Indeed, quercetin inhibits the growth, the adhesion and the migration of thyroid cancer cells, and it also has redifferentiation properties in some thyroid cancer cell lines. In conclusion, these data suggest that, although its effects can be of benefit in hyperthyroidism and thyroid cancer, caution is required in the use of high doses of quercetin due to its anti-thyroid properties. Further in vivo studies are certainly needed to confirm these hypotheses.
Collapse
Affiliation(s)
- Cesidio Giuliani
- Unit of Endocrinology, Department of Medicine and Sciences of Aging and Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy; (G.D.D.); (I.B.); (G.N.)
| | | | | | | |
Collapse
|
2
|
Liu G, Zhang CP, Lu YY, Niu DF, Hu FL. Biotransformation and metabolite activity analysis of flavonoids from propolis in vivo. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:1192-1206. [PMID: 38794953 DOI: 10.1080/10286020.2024.2355142] [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: 11/15/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024]
Abstract
Propolis is a natural resinous compound produced by bees, mixed with their saliva and wax, and has a range of biological benefits, including antioxidant and anti-inflammatory effects. This article reviews the in vivo transformation of propolis flavonoids and their potential influence on drug efficacy. Despite propolis is widely used, there is little research on how the active ingredients of propolis change in the body and how they interact with drugs. Future research will focus on these interactions and the metabolic fate of propolis in vivo.
Collapse
Affiliation(s)
- Gang Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cui-Ping Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuan-Yuan Lu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - De-Fang Niu
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Fu-Liang Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
3
|
Brodzicka A, Galanty A, Paśko P. Modulation of Multidrug Resistance Transporters by Food Components and Dietary Supplements: Implications for Cancer Therapy Efficacy and Safety. Curr Issues Mol Biol 2024; 46:9686-9706. [PMID: 39329928 PMCID: PMC11430623 DOI: 10.3390/cimb46090576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
The aim of this review is to explore how diet and dietary supplements influence the activity of key multidrug resistance (MDR) transporters-MRP2, BCRP, and P-gp. These transporters play a crucial role in drug efflux from cancer cells and significantly affect chemotherapy outcomes. This review focuses on how dietary phytochemicals, such as catechins and quercetin, impact the expression and function of these transporters. Both in vitro and in vivo experiments were examined to assess changes in drug bioavailability and intracellular drug accumulation. The findings show that certain dietary components-such as catechins, flavonoids, resveratrol, curcumin, terpenoids, sterols, and alkaloids-can either inhibit or induce MDR transporter activity, thus influencing the effectiveness of chemotherapy. These results highlight the importance of understanding diet-drug interactions in cancer therapy to improve treatment outcomes and reduce side effects. In conclusion, dietary modifications and supplements should be carefully considered in cancer treatment plans to optimize therapeutic efficacy.
Collapse
Affiliation(s)
- Agnieszka Brodzicka
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Agnieszka Galanty
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland;
| | - Paweł Paśko
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| |
Collapse
|
4
|
Carecho R, Marques D, Carregosa D, Masuero D, Garcia-Aloy M, Tramer F, Passamonti S, Vrhovsek U, Ventura MR, Brito MA, Nunes Dos Santos C, Figueira I. Circulating low-molecular-weight (poly)phenol metabolites in the brain: unveiling in vitro and in vivo blood-brain barrier transport. Food Funct 2024; 15:7812-7827. [PMID: 38967492 DOI: 10.1039/d4fo01396d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Circulating metabolites resulting from colonic metabolism of dietary (poly)phenols are highly abundant in the bloodstream, though still marginally explored, particularly concerning their brain accessibility. Our goal is to disclose (poly)phenol metabolites' blood-brain barrier (BBB) transport, in vivo and in vitro, as well as their role at BBB level. For three selected metabolites, benzene-1,2-diol-3-sulfate/benzene-1,3-diol-2-sulfate (pyrogallol-sulfate - Pyr-sulf), benzene-1,3-diol-6-sulfate (phloroglucinol-sulfate - Phlo-sulf), and phenol-3-sulfate (resorcinol-sulfate - Res-sulf), BBB transport was assessed in human brain microvascular endothelial cells (HBMEC). Their potential in modulating in vitro BBB properties at circulating concentrations was also studied. Metabolites' fate towards the brain, liver, kidney, urine, and blood was disclosed in Wistar rats upon injection. Transport kinetics in HBMEC highlighted different BBB permeability rates, where Pyr-sulf emerged as the most in vitro BBB permeable metabolite. Pyr-sulf was also the most potent regarding BBB properties improvement, namely increased beta(β)-catenin membrane expression and reduction of zonula occludens-1 membrane gaps. Whereas no differences were observed for transferrin, increased expression of caveolin-1 upon Pyr-sulf and Res-sulf treatments was found. Pyr-sulf was also capable of modulating gene and protein expression of some solute carrier transporters. Notably, each of the injected metabolites exhibited a unique tissue distribution in vivo, with the remarkable ability to almost immediately reach the brain.
Collapse
Affiliation(s)
- Rafael Carecho
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Avenida da República, Oeiras, Portugal
| | - Daniela Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
| | - Diogo Carregosa
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
| | - Domenico Masuero
- Metabolomics Unit, Research and Innovation Centre, Fondazione Edmund Mach (FEM), via E. Mach 1, San Michele all'Adige, Italy
| | - Mar Garcia-Aloy
- Metabolomics Unit, Research and Innovation Centre, Fondazione Edmund Mach (FEM), via E. Mach 1, San Michele all'Adige, Italy
| | - Federica Tramer
- Department of Life Sciences, University of Trieste, via L. Giorgieri 1, Trieste, Italy
| | - Sabina Passamonti
- Department of Life Sciences, University of Trieste, via L. Giorgieri 1, Trieste, Italy
| | - Urska Vrhovsek
- Metabolomics Unit, Research and Innovation Centre, Fondazione Edmund Mach (FEM), via E. Mach 1, San Michele all'Adige, Italy
| | - M Rita Ventura
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Avenida da República, Oeiras, Portugal
| | - Maria Alexandra Brito
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, Portugal
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Avenida da República, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Avenida da República, Apartado 12, Oeiras, Portugal
| | - Inês Figueira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
| |
Collapse
|
5
|
Wang FH, Tan HX, Hu JH, Duan XY, Bai WT, Wang XB, Wang BL, Su Y, Hu JP. Inhibitory interaction of flavonoids with organic anion transporter 3 and their structure-activity relationships for predicting nephroprotective effects. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:353-371. [PMID: 37589480 DOI: 10.1080/10286020.2023.2240722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/20/2023] [Indexed: 08/18/2023]
Abstract
The organic anion transporter 3 (OAT3), an important renal uptake transporter, is associated with drug-induced acute kidney injury (AKI). Screening and identifying potent OAT3 inhibitors with little toxicity in natural products, especially flavonoids, in reducing OAT3-mediated AKI is of great value. The five strongest OAT3 inhibitors from the 97 flavonoids markedly decreased aristolochic acid I-induced cytotoxicity and alleviated methotrexate-induced nephrotoxicity. The pharmacophore model clarified hydrogen bond acceptors and hydrophobic groups are the critical pharmacophores. These findings would provide valuable information in predicting the potential risks of flavonoid-containing food/herb-drug interactions and optimizing flavonoid structure to alleviate OAT3-related AKI.
Collapse
Affiliation(s)
- Feng-He Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hui-Xin Tan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jia-Huan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Department of Health Management and Service, Cangzhou Medical College, Cangzhou 061001, China
| | - Xiao-Yan Duan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wan-Ting Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xin-Bo Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bao-Lian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yan Su
- Department of Health Management and Service, Cangzhou Medical College, Cangzhou 061001, China
| | - Jin-Ping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
6
|
Yu S, Zheng J, Zhang Y, Meng D, Wang Y, Xu X, Liang N, Shabiti S, Zhang X, Wang Z, Yang Z, Mi P, Zheng X, Li W, Chen H. The mechanisms of multidrug resistance of breast cancer and research progress on related reversal agents. Bioorg Med Chem 2023; 95:117486. [PMID: 37847948 DOI: 10.1016/j.bmc.2023.117486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/18/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023]
Abstract
Chemotherapy is the mainstay in the treatment of breast cancer. However, many drugs that are commonly used in clinical practice have a high incidence of side effects and multidrug resistance (MDR), which is mainly caused by overexpression of drug transporters and related enzymes in breast cancer cells. In recent years, researchers have been working hard to find newer and safer drugs to overcome MDR in breast cancer. In this review, we provide the molecule mechanism of MDR in breast cancer, categorize potential lead compounds that inhibit single or multiple drug transporter proteins, as well as related enzymes. Additionally, we have summarized the structure-activity relationship (SAR) based on potential breast cancer MDR modulators with lower side effects. The development of novel approaches to suppress MDR is also addressed. These lead compounds hold great promise for exploring effective chemotherapy agents to overcome MDR, providing opportunities for curing breast cancer in the future.
Collapse
Affiliation(s)
- Shiwen Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Jinling Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Yan Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Dandan Meng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Yujue Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Xiaoyu Xu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Na Liang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Shayibai Shabiti
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Xu Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zixi Wang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zehua Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Pengbing Mi
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Xing Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Department of Pharmacy, Hunan Vocational College of Science and Technology, Third Zhongyi Shan Road, Changsha, Hunan Province 425101, PR China.
| | - Wenjun Li
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Hongfei Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China.
| |
Collapse
|
7
|
Zargar S, Altwaijry N, Wani TA, Alkahtani HM. Evaluation of the Possible Pathways Involved in the Protective Effects of Quercetin, Naringenin, and Rutin at the Gene, Protein and miRNA Levels Using In-Silico Multidimensional Data Analysis. Molecules 2023; 28:4904. [PMID: 37446564 DOI: 10.3390/molecules28134904] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Flavonoids are secondary metabolites that are non-essential for plant growth or survival, and they also provide numerous health benefits to humans. They are antioxidants that shield plants from the ill effects of ultraviolet light, pests, and diseases. They are beneficial to health for several reasons, including lowering inflammation, boosting cardiovascular health, and lowering cancer risk. This study looked into the physicochemical features of these substances to determine the potential pharmacological pathways involved in their protective actions. Potential targets responsible for the protective effects of quercetin, naringenin, and rutin were identified with SwissADME. The associated biological processes and protein-protein networks were analyzed by using the GeneMANIA, Metascape, and STRING servers. All the flavonoids were predicted to be orally bioavailable, with more than 90% targets as enzymes, including kinases and lyases, and with common targets such as NOS2, CASP3, CASP9, CAT, BCL2, TNF, and HMOX1. TNF was shown to be a major target in over 250 interactions. To extract the "biological meanings" from the MCODE networks' constituent parts, a GO enrichment analysis was performed on each one. The most important transcription factors in gene regulation were RELA, NFKB1, PPARG, and SP1. Treatment with quercetin, naringenin, or rutin increased the expression and interaction of the microRNAs' hsa-miR-34a-5p, hsa-miR-30b-5p, hsa-let-7a-5p, and hsa-miR-26a-1-3p. The anticancer effects of hsa-miR-34a-5p have been experimentally confirmed. It also plays a critical role in controlling other cancer-related processes such as cell proliferation, apoptosis, EMT, and metastasis. This study's findings might lead to a deeper comprehension of the mechanisms responsible for flavonoids' protective effects and could present new avenues for exploration.
Collapse
Affiliation(s)
- Seema Zargar
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Tanveer A Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
8
|
Liu Y, Nie X, Wang J, Zhao Z, Wang Z, Ju F. Visualizing the distribution of flavonoids in litchi ( Litchi chinenis) seeds through matrix-assisted laser desorption/ionization mass spectrometry imaging. FRONTIERS IN PLANT SCIENCE 2023; 14:1144449. [PMID: 36909412 PMCID: PMC9998689 DOI: 10.3389/fpls.2023.1144449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Flavonoids are one of the most important bioactive components in litchi (Litchi chinensis Sonn.) seeds and have broad-spectrum antiviral and antitumor activities. Litchi seeds have been shown to inhibit the proliferation of cancer cells and induce apoptosis, particularly effective against breast and liver cancers. Elucidating the distribution of flavonoids is important for understanding their physiological and biochemical functions and facilitating their efficient extraction and utilization. However, the spatial distribution patterns and expression states of flavonoids in litchi seeds remain unclear. Herein, matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) was used for in situ detection and imaging of the distribution of flavonoids in litchi seed tissue sections for the first time. Fifteen flavonoid ion signals, including liquiritigenin, apigenin, naringenin, luteolin, dihydrokaempferol, daidzein, quercetin, taxifolin, kaempferol, isorhamnetin, myricetin, catechin, quercetin 3-β-d-glucoside, baicalin, and rutin, were successfully detected and imaged in situ through MALDI-MSI in the positive ion mode using 2-mercaptobenzothiazole as a matrix. The results clearly showed the heterogeneous distribution of flavonoids, indicating the potential of litchi seeds for flavonoid compound extraction. MALDI-MS-based multi-imaging enhanced the visualization of spatial distribution and expression states of flavonoids. Thus, apart from improving our understanding of the spatial distribution of flavonoids in litchi seeds, our findings also facilitate the development of MALDI-MSI-based metabolomics as a novel effective molecular imaging tool for evaluating the spatial distribution of endogenous compounds.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Breast Surgery, Breast Disease Center, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Xiaofei Nie
- Department of Oncology, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Jilong Wang
- Department of Acupuncture and Moxibustion, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Zhenqi Zhao
- Department of Radiology, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Zhimei Wang
- Department of Gynecological Neoplasms, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Fang Ju
- Department of Oncology, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Mao X, Li H, Zheng J. Effects of xenobiotics on CYP1 enzyme-mediated biotransformation and bioactivation of estradiol. Drug Metab Rev 2023; 55:1-49. [PMID: 36823774 DOI: 10.1080/03602532.2023.2177671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Endogenous estradiol (E2) exerts diverse physiological and pharmacological activities, commonly used for hormone replacement therapy. However, prolonged and excessive exposure to E2 potentially increases estrogenic cancer risk. Reportedly, CYP1 enzyme-mediated biotransformation of E2 is largely concerned with its balance between detoxification and carcinogenic pathways. Among the three key CYP1 enzymes (CYP1A1, CYP1A2, and CYP1B1), CYP1A1 and CYP1A2 mainly catalyze the formation of nontoxic 2-hydroxyestradiol (2-OH-E2), while CYP1B1 specifically catalyzes the formation of genotoxic 4-hydroxyestradiol (4-OH-E2). 4-OH-E2 can be further metabolized to electrophilic quinone intermediates accompanied by the generation of reactive oxygen species (ROS), triggering DNA damage. Since abnormal alterations in CYP1 activities can greatly affect the bioactivation process of E2, regulatory effects of xenobiotics on CYP1s are essential for E2-associated cancer development. To date, thousands of natural and synthetic compounds have been found to show potential inhibition and/or induction actions on the three CYP1 members. Generally, these chemicals share similar planar polycyclic skeletons, the structural motifs and substituent groups of which are important for their inhibitory/inductive efficiency and selectivity toward CYP1 enzymes. This review comprehensively summarizes these known inhibitors and/or inductors of E2-metabolizing CYP1s based on chemical categories and discusses their structure-activity relationships, which would contribute to better understanding of the correlation between xenobiotic-regulated CYP1 activities and estrogenic cancer susceptibility.
Collapse
Affiliation(s)
- Xu Mao
- Department of Pharmaceutical Analysis, College of Pharmacy, Mudanjiang Medical University, Mudanjiang, China
| | - Hui Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| |
Collapse
|
10
|
Improvement of Therapeutic Value of Quercetin with Chitosan Nanoparticle Delivery Systems and Potential Applications. Int J Mol Sci 2023; 24:ijms24043293. [PMID: 36834702 PMCID: PMC9959398 DOI: 10.3390/ijms24043293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
This paper reviews recent studies investigating chitosan nanoparticles as drug delivery systems for quercetin. The therapeutic properties of quercetin include antioxidant, antibacterial and anti-cancer potential, but its therapeutic value is limited by its hydrophobic nature, low bioavailability and fast metabolism. Quercetin may also act synergistically with other stronger drugs for specific disease states. The encapsulation of quercetin in nanoparticles may increase its therapeutic value. Chitosan nanoparticles are a popular candidate in preliminary research, but the complex nature of chitosan makes standardisation difficult. Recent studies have used in-vitro, and in-vivo experiments to study the delivery of quercetin alone or in combination with another active pharmaceutical ingredient encapsulated in chitosan nanoparticles. These studies were compared with the administration of non-encapsulated quercetin formulation. Results suggest that encapsulated nanoparticle formulations are better. In-vivo or animal models simulated the type of disease required to be treated. The types of diseases were breast, lung, liver and colon cancers, mechanical and UVB-induced skin damage, cataracts and general oxidative stress. The reviewed studies included various routes of administration: oral, intravenous and transdermal routes. Although toxicity tests were often included, it is believed that the toxicity of loaded nanoparticles needs to be further researched, especially when not orally administered.
Collapse
|
11
|
Manthalkar L, Ajazuddin, Bhattacharya S. Evidence-based capacity of natural cytochrome enzyme inhibitors to increase the effectivity of antineoplastic drugs. Discov Oncol 2022; 13:142. [PMID: 36571647 PMCID: PMC9792636 DOI: 10.1007/s12672-022-00605-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/14/2022] [Indexed: 12/27/2022] Open
Abstract
Cytochrome (CYP) enzymes catalyze the metabolism of numerous exogenous and endogenous substrates in cancer therapy leading to significant drug interactions due to their metabolizing effect. CYP enzymes play an important role in the metabolism of essential anticancer medications. They are shown to be overexpressed in tumor cells at numerous locations in the body. This overexpression could be a result of lifestyle factors, presence of hereditary variants of CYP (Bio individuality) and multi-drug resistance. This finding has sparked an interest in using CYP inhibitors to lower their metabolizing activity as a result facilitating anti-cancer medications to have a therapeutic impact. As a result of the cytotoxic nature of synthetic enzyme inhibitors and the increased prevalence of herbal medication, natural CYP inhibitors have been identified as an excellent way to inhibit overexpression sighting their tendency to show less cytotoxicity, lesser adverse drug reactions and enhanced bioavailability. Nonetheless, their effect of lowering the hindrance caused in chemotherapy due to CYP enzymes remains unexploited to its fullest. It has been observed that there is a substantial decrease in first pass metabolism and increase in intestinal absorption of chemotherapeutic drugs like paclitaxel when administered along with flavonoids which help suppress certain specific cytochrome enzymes which play a role in paclitaxel metabolism. This review elaborates on the role and scope of phytochemicals in primary, secondary and tertiary care and how targeted prevention of cancer could be a breakthrough in the field of chemotherapy and oncology. This opens up a whole new area of research for delivery of these natural inhibitors along with anticancer drugs with the help of liposomes, micelles, nanoparticles, the usage of liquid biopsy analysis, artificial intelligence in medicine, risk assessment tools, multi-omics and multi-parametric analysis. Further, the site of action, mechanisms, metabolites involved, experimental models, doses and observations of two natural compounds, quercetin & thymoquinone, and two plant extracts, liquorice & garlic on CYP enzymes have been summarized.
Collapse
Affiliation(s)
- Laxmi Manthalkar
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, 425405, Maharashtra, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences & Research, Khoka-Kurud Road, Bhilai, 490024, Chhattisgarh, India.
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, 425405, Maharashtra, India.
| |
Collapse
|
12
|
Recent Advances in Natural Polyphenol Research. Molecules 2022; 27:molecules27248777. [PMID: 36557912 PMCID: PMC9787743 DOI: 10.3390/molecules27248777] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Polyphenols are secondary metabolites produced by plants, which contribute to the plant's defense against abiotic stress conditions (e.g., UV radiation and precipitation), the aggression of herbivores, and plant pathogens. Epidemiological studies suggest that long-term consumption of plant polyphenols protects against cardiovascular disease, cancer, osteoporosis, diabetes, and neurodegenerative diseases. Their structural diversity has fascinated and confronted analytical chemists on how to carry out unambiguous identification, exhaustive recovery from plants and organic waste, and define their nutritional and biological potential. The food, cosmetic, and pharmaceutical industries employ polyphenols from fruits and vegetables to produce additives, additional foods, and supplements. In some cases, nanocarriers have been used to protect polyphenols during food processing, to solve the issues related to low water solubility, to transport them to the site of action, and improve their bioavailability. This review summarizes the structure-bioactivity relationships, processing parameters that impact polyphenol stability and bioavailability, the research progress in nanocarrier delivery, and the most innovative methodologies for the exhaustive recovery of polyphenols from plant and agri-waste materials.
Collapse
|
13
|
Pei S, Dou Y, Zhang W, Qi D, Li Y, Wang M, Li W, Shi H, Gao Z, Yao C, Fang D, Sun H, Xie S. O-Sulfation disposition of curcumin and quercetin in SULT1A3 overexpressing HEK293 cells: the role of arylsulfatase B in cellular O-sulfation regulated by transporters. Food Funct 2022; 13:10558-10573. [PMID: 36156668 DOI: 10.1039/d2fo01436j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Extensive phase II metabolic reactions (i.e., glucuronidation and sulfation) have resulted in low bioavailability and decreased biological effects of curcumin and quercetin. Compared to glucuronidation, information on the sulfation disposition of curcumin and quercetin is limited. In this study, we identified that BCRP and MRP4 played a critical role in the cellular excretion of curcumin-O-sulfate (C-O-S) and quercetin-O-sulfate (Q-O-S) by integrating chemical inhibition with transporter knock-down experiments. Inhibited excretion of sulfate (C-O-S and Q-O-S) caused significant reductions in cellular O-sulfation of curcumin (a maximal 74.4% reduction) and quercetin (a maximal 76.9% reduction), revealing a strong interplay of sulfation with efflux transport. It was further identified that arylsulfatase B (ARSB) played a crucial role in the regulation of cellular O-sulfation by transporters. ARSB overexpression significantly enhanced the reduction effect of MK-571 on the cellular O-sulfation (fmet) of the model compound (38.8% reduction for curcumin and 44.2% reduction for quercetin). On the contrary, ARSB knockdown could reverse the effect of MK-571 on the O-sulfation disposition of the model compound (29.7% increase for curcumin and 47.3% increase for quercetin). Taken together, ARSB has been proven to be involved in cellular O-sulfation, accounting for transporter-dependent O-sulfation of curcumin and quercetin. A better understanding of the interplay beneath metabolism and transport will contribute to the exact prediction of in vivo drug disposition and drug-drug interactions.
Collapse
Affiliation(s)
- Shuhua Pei
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Yuanyuan Dou
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Wenke Zhang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Defei Qi
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Yingying Li
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Mengqing Wang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Wenqi Li
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Hongxiang Shi
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Zixuan Gao
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Chaoyan Yao
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Dong Fang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China. .,Academy for advanced interdisciplinary studies, Henan University, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Hua Sun
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China. .,Academy for advanced interdisciplinary studies, Henan University, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Songqiang Xie
- Academy for advanced interdisciplinary studies, Henan University, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China. .,Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| |
Collapse
|
14
|
Muscogiuri G, Barrea L, Cantone MC, Guarnotta V, Mazzilli R, Verde L, Vetrani C, Colao A, Faggiano A. Neuroendocrine Tumors: A Comprehensive Review on Nutritional Approaches. Cancers (Basel) 2022; 14:cancers14184402. [PMID: 36139562 PMCID: PMC9496842 DOI: 10.3390/cancers14184402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Neuroendocrine neoplasms are a heterogeneous group of neoplasms with increasing incidence, high prevalence, and survival worldwide. About 90% of cases are well differentiated forms, the so-called neuroendocrine tumors (NETs), with slow proliferation rates and prolonged survival but frequent development of liver metastases and endocrine syndromes. Both the tumor itself and systemic therapy may have an impact on patient nutrition. Malnutrition has a negative impact on outcome in patients with NETs, as well as obesity. In addition, obesity and metabolic syndrome have been shown to be risk factors for both the development and prognosis of NET. Therefore, dietary assessment based on body composition and lifestyle modifications should be an integral part of the treatment of NET patients. Nutrition plans, properly formulated by a dietician, are an integral part of the multidisciplinary treatment team for patients with NETs because they allow an improvement in quality of life, providing a tailored approach based on nutritional needs and nutritional manageable signs and/or symptoms related to pharmacological treatment. The aim of this review is to condense the latest evidence on the role of the most used dietary models, the Mediterranean diet, the ketogenic diet, and intermittent fasting, in the context of NETs, while considering the clinical and molecular mechanisms by which these dietary models act.
Collapse
Affiliation(s)
- Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
- Unità di Endocrinologia, Diabetologia ed Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
- Cattedra Unesco “Educazione alla Salute e allo Sviluppo Sostenibile”, Università Federico II, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-0817463779; Fax: +39-081-746-3688
| | - Luigi Barrea
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, 80143 Naples, Italy
| | - Maria Celeste Cantone
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, 20157 Milan, Italy
| | - Valentina Guarnotta
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro” (PROMISE), Sezione di Malattie Endocrine, del Ricambio e della Nutrizione, Università di Palermo, 90127 Palermo, Italy
| | - Rossella Mazzilli
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00185 Rome, Italy
| | - Ludovica Verde
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
| | - Claudia Vetrani
- Unità di Endocrinologia, Diabetologia ed Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
- Cattedra Unesco “Educazione alla Salute e allo Sviluppo Sostenibile”, Università Federico II, 80131 Naples, Italy
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
- Unità di Endocrinologia, Diabetologia ed Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università Federico II, 80131 Naples, Italy
- Cattedra Unesco “Educazione alla Salute e allo Sviluppo Sostenibile”, Università Federico II, 80131 Naples, Italy
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
15
|
Hildreth SB, Littleton ES, Clark LC, Puller GC, Kojima S, Winkel BSJ. Mutations that alter Arabidopsis flavonoid metabolism affect the circadian clock. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2022; 110:932-945. [PMID: 35218268 PMCID: PMC9311810 DOI: 10.1111/tpj.15718] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 05/05/2023]
Abstract
Flavonoids are a well-known class of specialized metabolites that play key roles in plant development, reproduction, and survival. Flavonoids are also of considerable interest from the perspective of human health, as both phytonutrients and pharmaceuticals. RNA sequencing analysis of an Arabidopsis null allele for chalcone synthase (CHS), which catalyzes the first step in flavonoid metabolism, has uncovered evidence that these compounds influence the expression of genes associated with the plant circadian clock. Analysis of promoter-luciferase constructs further showed that the transcriptional activity of CCA1 and TOC1, two key clock genes, is altered in CHS-deficient seedlings across the day/night cycle. Similar findings for a mutant line lacking flavonoid 3'-hydroxylase (F3'H) activity, and thus able to synthesize mono- but not dihydroxylated B-ring flavonoids, suggests that the latter are at least partially responsible; this was further supported by the ability of quercetin to enhance CCA1 promoter activity in wild-type and CHS-deficient seedlings. The effects of flavonoids on circadian function were also reflected in photosynthetic activity, with chlorophyll cycling abolished in CHS- and F3'H-deficient plants. Remarkably, the same phenotype was exhibited by plants with artificially high flavonoid levels, indicating that neither the antioxidant potential nor the light-screening properties of flavonoids contribute to optimal clock function, as has recently also been demonstrated in animal systems. Collectively, the current experiments point to a previously unknown connection between flavonoids and circadian cycling in plants and open the way to better understanding of the molecular basis of flavonoid action.
Collapse
Affiliation(s)
- Sherry B. Hildreth
- Department of Biological SciencesVirginia TechBlacksburgVA24061USA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVA24061USA
| | - Evan S. Littleton
- Department of Biological SciencesVirginia TechBlacksburgVA24061USA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVA24061USA
| | - Leor C. Clark
- Department of Biological SciencesVirginia TechBlacksburgVA24061USA
- Present address:
Department of Global Health, Milken Institute School of Public HealthGeorge Washington UniversityWashingtonDC20052USA
| | - Gabrielle C. Puller
- Department of Biological SciencesVirginia TechBlacksburgVA24061USA
- Present address:
Laboratory of Molecular BiologyNational Cancer InstituteNational Institutes of HealthBethesdaMD20 892USA
| | - Shihoko Kojima
- Department of Biological SciencesVirginia TechBlacksburgVA24061USA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVA24061USA
| | - Brenda S. J. Winkel
- Department of Biological SciencesVirginia TechBlacksburgVA24061USA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVA24061USA
| |
Collapse
|
16
|
Nutmakul T. A review on benefits of quercetin in hyperuricemia and gouty arthritis. Saudi Pharm J 2022; 30:918-926. [PMID: 35903522 PMCID: PMC9315272 DOI: 10.1016/j.jsps.2022.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Hyperuricemia becomes a public health problem worldwide. It is not only a major risk factor for gout but also associated with the development of life-threatening diseases such as chronic kidney disease and cardiovascular diseases. Although there are several available therapeutic drugs, some serious adverse effects and contraindications are concerned. These drive the search for an alternative therapy that is effective and safe. Quercetin is of particular interesting since it has been reported numerous pharmacological activities, especially anti-hyperuricemia, antioxidant, anti-inflammation and amelioration of metabolic syndromes and cardiovascular diseases which are comorbidities of hyperuricemia and gout. In addition, quercetin has been widely used as a health supplement for many diseases however, the use for hyperuricemia and gout has not been indicated. Therefore, this review aims to gather and summarize published data regarding the efficacy in preclinical and clinical studies along with possible mechanism of action, and safety aspect of quercetin in order to support the use of quercetin as a dietary supplement for prevention and management of hyperuricemia and gouty arthritis and/or use as alternative or combination therapy to minimize the side effects of the conventional drugs.
Collapse
|
17
|
Li J, Wu Y, Ma Y, Bai L, Li Q, Zhou X, Xu P, Li X, Xue M. A UPLC-MS/MS method reveals the pharmacokinetics and metabolism characteristics of kaempferol in rats under hypoxia. Drug Metab Pharmacokinet 2022; 43:100440. [PMID: 35051732 DOI: 10.1016/j.dmpk.2021.100440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/24/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022]
|
18
|
Britza SM, Musgrave IF, Byard RW. Implications for herbal polypharmacy: coumarin-induced hepatotoxicity increased through common herbal phytochemicals astragaloside IV and atractylenolide I. Toxicol Mech Methods 2022; 32:606-615. [PMID: 35354423 DOI: 10.1080/15376516.2022.2057267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hepatotoxicity is a well-known adverse effect of many substances, with toxicity often resulting from interactions of drugs with other drug-like substances. With the increased availability of complementary and alternative medicines, including herbal medicines, the likelihood of adverse interactions between drugs and drug-like substances in herbs increases. However, the impact of potential herb-herb interactions is little understood. To assess the potential of two cytochrome P450 enzyme modulating phytochemicals common to many herbal medicines, atractylenolide I (ATR-I) and astragaloside IV (AST-IV), to interact with coumarin, another phytochemical common in many foods, a hepatocyte function model with a liver carcinoma cell line, HepG2, was exposed to these agents. To determine the effects of cytochrome P450 modulation by these phytochemicals certain cells were induced with rifampicin to induce cytochrome P450. Increasing concentrations of ATR-I combined with a fixed, nontoxic concentration of coumarin (200 µM), demonstrated significant additive interactions. 300 µM ATR-I produced a 31% reduction in cell viability (p < 0.01) with coumarin in rifampicin uninduced cells. In rifampicin-induced cells, ATR-I (100-300 µM) produced a significant reduction in cell viability (p < 0.01) with coumarin (200 µM). AST-IV with fixed coumarin (200 µM) showed 27% toxicity at 300 µM AST-IV in rifampicin uninduced cells (p < 0.05) and 30% toxicity in rifampicin induced cells (p < 0.05). However, when fixed coumarin and AST-IV were combined with increasing concentrations of ATR-I no further significant increase in toxicity was observed (p > 0.05). These results demonstrate the potential toxic interactive capabilities of common traditional Chinese herbal medicine phytochemicals and underline the potential importance of coumarin-mediated toxicity.
Collapse
Affiliation(s)
- Susan M Britza
- School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Ian F Musgrave
- School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Roger W Byard
- School of Biomedicine, The University of Adelaide, Adelaide, Australia.,Forensic Science South Australia, Adelaide, Australia
| |
Collapse
|
19
|
Fatima N, Baqri SSR, Bhattacharya A, Koney NKK, Husain K, Abbas A, Ansari RA. Role of Flavonoids as Epigenetic Modulators in Cancer Prevention and Therapy. Front Genet 2021; 12:758733. [PMID: 34858475 PMCID: PMC8630677 DOI: 10.3389/fgene.2021.758733] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022] Open
Abstract
Epigenetic regulation involves reversible changes in histones and DNA modifications that can be inherited without any changes in the DNA sequence. Dysregulation of normal epigenetic processes can lead to aberrant gene expression as observed in many diseases, notably cancer. Recent insights into the mechanisms of DNA methylation, histone modifications, and non-coding RNAs involved in altered gene expression profiles of tumor cells have caused a paradigm shift in the diagnostic and therapeutic approaches towards cancer. There has been a surge in search for compounds that could modulate the altered epigenetic landscape of tumor cells, and to exploit their therapeutic potential against cancers. Flavonoids are naturally occurring phenol compounds which are abundantly found among phytochemicals and have potentials to modulate epigenetic processes. Knowledge of the precise flavonoid-mediated epigenetic alterations is needed for the development of epigenetics drugs and combinatorial therapeutic approaches against cancers. This review is aimed to comprehensively explore the epigenetic modulations of flavonoids and their anti-tumor activities.
Collapse
Affiliation(s)
- Nishat Fatima
- Department of Chemistry, Shia Postgraduate College, Lucknow, India
| | | | - Atrayee Bhattacharya
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Nii Koney-Kwaku Koney
- Department of Anatomy, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Kazim Husain
- Department of Molecular Medicine, University of South Florida, Tampa, FL, United States
| | - Ata Abbas
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Rais A Ansari
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, United States
| |
Collapse
|
20
|
Iglesias-Aguirre CE, Cortés-Martín A, Ávila-Gálvez MÁ, Giménez-Bastida JA, Selma MV, González-Sarrías A, Espín JC. Main drivers of (poly)phenol effects on human health: metabolite production and/or gut microbiota-associated metabotypes? Food Funct 2021; 12:10324-10355. [PMID: 34558584 DOI: 10.1039/d1fo02033a] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the high human interindividual variability in response to (poly)phenol consumption, the cause-and-effect relationship between some dietary (poly)phenols (flavanols and olive oil phenolics) and health effects (endothelial function and prevention of LDL oxidation, respectively) has been well established. Most of the variables affecting this interindividual variability have been identified (food matrix, gut microbiota, single-nucleotide-polymorphisms, etc.). However, the final drivers for the health effects of (poly)phenol consumption have not been fully identified. At least partially, these drivers could be (i) the (poly)phenols ingested that exert their effect in the gastrointestinal tract, (ii) the bioavailable metabolites that exert their effects systemically and/or (iii) the gut microbial ecology associated with (poly)phenol metabolism (i.e., gut microbiota-associated metabotypes). However, statistical associations between health effects and the occurrence of circulating and/or excreted metabolites, as well as cross-sectional studies that correlate gut microbial ecologies and health, do not prove a causal role unequivocally. We provide a critical overview and perspective on the possible main drivers of the effects of (poly)phenols on human health and suggest possible actions to identify the putative actors responsible for the effects.
Collapse
Affiliation(s)
- Carlos E Iglesias-Aguirre
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain.
| | - Adrián Cortés-Martín
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain.
| | - María Á Ávila-Gálvez
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal.,Instituto de Biologia Experimental e Tecnológica (IBET), Apartado 12, 2781-901, Oeiras, Portugal
| | - Juan A Giménez-Bastida
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain.
| | - María V Selma
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain.
| | - Antonio González-Sarrías
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain.
| | - Juan Carlos Espín
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, 30100 Campus de Espinardo, Murcia, Spain.
| |
Collapse
|
21
|
Topoisomerase poisoning by the flavonoid nevadensin triggers DNA damage and apoptosis in human colon carcinoma HT29 cells. Arch Toxicol 2021; 95:3787-3802. [PMID: 34635930 PMCID: PMC8536574 DOI: 10.1007/s00204-021-03162-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022]
Abstract
Nevadensin, an abundant polyphenol of basil, is reported to reduce alkenylbenzene DNA adduct formation. Furthermore, it has a wide spectrum of further pharmacological properties. The presented study focuses the impact of nevadensin on topoisomerases (TOPO) in vitro. Considering the DNA-intercalating properties of flavonoids, first, minor groove binding properties (IC50 = 31.63 µM), as well as DNA intercalation (IC50 = 296.91 µM) of nevadensin, was found. To determine potential in vitro effects on TOPO I and TOPO IIα, the relaxation and decatenation assay was performed in a concentration range of 1–500 µM nevadensin. A partial inhibition was detected for TOPO I at concentrations ≥ 100 µM, whereas TOPO IIα activity is only inhibited at concentrations ≥ 250 µM. To clarify the mode of action, the isolating in vivo complex of enzyme assay was carried out using human colon carcinoma HT29 cells. After 1 h of incubation, the amount of TOPO I linked to DNA was significantly increased by nevadensin (500 µM), why nevadensin was characterized as TOPO I poison. However, no effects on TOPO IIα were detected in the cellular test system. As a subsequent cellular response to TOPO I poisoning, a highly significant increase of DNA damage after 2 h and a decrease of cell viability after 48 h at the same concentration range were found. Furthermore, after 24 h of incubation a G2/M arrest was observed at concentrations ≥ 100 µM by flow cytometry. The analysis of cell death revealed that nevadensin induces the intrinsic apoptotic pathway via activation of caspase-9 and caspase-3. The results suggest that cell cycle disruption and apoptotic events play key roles in the cellular response to TOPO I poisoning caused by nevadensin in HT29 cells.
Collapse
|
22
|
Elsbaey M, Ibrahim MAA, Bar FA, Elgazar AA. Chemical constituents from coconut waste and their in silico evaluation as potential antiviral agents against SARS-CoV-2. SOUTH AFRICAN JOURNAL OF BOTANY : OFFICIAL JOURNAL OF THE SOUTH AFRICAN ASSOCIATION OF BOTANISTS = SUID-AFRIKAANSE TYDSKRIF VIR PLANTKUNDE : AMPTELIKE TYDSKRIF VAN DIE SUID-AFRIKAANSE GENOOTSKAP VAN PLANTKUNDIGES 2021; 141:278-289. [PMID: 34092840 PMCID: PMC8162769 DOI: 10.1016/j.sajb.2021.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/28/2021] [Accepted: 05/22/2021] [Indexed: 05/10/2023]
Abstract
Eleven compounds were isolated from the ethyl acetate extract of Cocos nucifera L endocarp, jezonofol 1, scirpusin A 2, cassigarol G 3, maackin A 4, threoguiacyl glycerol-8'-vanillic acid ether 5, erythroguiacyl glycerol-8'-vanillic acid ether 6, apigenin-7-O-β-D-glucoside 7, piceatannol 8, p-hydroxy-benzoic acid 9, protocatechuic acid 10 and vanillic acid 11. Compounds 1-7 were isolated for the first time from the plant. The isolated compounds were virtually screened against four critical components of severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), the main protease (Mpro), papain-like protease (PLpro), nonstructural protein 13 (nsp13) and RNA dependent RNA polymerase (RdRp). Stilbene dimers 1-4 showed remarkable binding affinities towards the investigated targets (binding energy <-7.6 kcal/mol). Compounds 1, 3 and 4 interacted with the catalytic dyad (Cys145-His41) at the active pocket of Mpro which is essential for achieving good inhibitory activity. Compounds 1-3 showed molecular interaction with the conserved ubiquitin-specific protease residues of PLpro, responsible for binding ability at different active sites of nsp13, which are crucial for decreasing the resistance caused by viral immune evasion. Compounds 2 and 3 showed the ability to bind at different active sites of nsp13, which is a key binding site for reducing antiviral resistance. Finally, compounds 1-3 showed the ability to bind with RdRp before and after RNA binding. Our findings suggested that the dimeric stilbene skeleton is a promising candidate for developing anti-COVID-19 drugs. Particularly, 1, 2 and 3, showed a promiscuity pattern binding to multiple targets of SARS-CoV-2 replication. Herein, 20 ns molecular dynamics (MD) simulations combined with molecular mechanics-generalized Born surface area (MM-GBSA) binding energy calculations were performed to estimate the binding affinity of the most potent three compounds against the viral SARS-CoV-2 targets. MM-GBSA calculations unveiled the outshine potency of compound 1 towards PLpro with a binding energy of -60.7 kcal/mol. Structural and energetic analyses over 20 ns MD simulation displayed the high stability of compound 1 in complex with PLpro. The list of the compounds was considered herein forms a primer for clinical investigation in COVID-19 patients and directing for further antiviral examinations. Drug likeness properties of compounds 1-4 were evaluated.
Collapse
Affiliation(s)
- Marwa Elsbaey
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, 35516, Egypt
| | - Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, 61519, Egypt
| | - Fatma Abdel Bar
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Faculty of Pharmacy, Mansoura University, 35516, Egypt
| | - Abdullah A Elgazar
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, 33516, Egypt
| |
Collapse
|
23
|
Bungsu I, Kifli N, Ahmad SR, Ghani H, Cunningham AC. Herbal Plants: The Role of AhR in Mediating Immunomodulation. Front Immunol 2021; 12:697663. [PMID: 34249001 PMCID: PMC8264659 DOI: 10.3389/fimmu.2021.697663] [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] [Received: 04/20/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022] Open
Abstract
The prevalence of chronic inflammatory diseases including inflammatory bowel disease (IBD), autoimmunity and cancer have increased in recent years. Herbal-based compounds such as flavonoids have been demonstrated to contribute to the modulation of these diseases although understanding their mechanism of action remains limited. Flavonoids are able to interact with cellular immune components in a distinct way and influence immune responses at a molecular level. In this mini review, we highlight recent progress in our understanding of the modulation of immune responses by the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor whose activity can be regulated by diverse molecules including flavonoids. We focus on the role of AhR in integrating signals from flavonoids to modulate inflammatory responses using in vitro and experimental animal models. We also summarize the limitations of these studies. Medicinal herbs have been widely used to treat inflammatory disorders and may offer a valuable therapeutic strategy to treat aberrant inflammatory responses by modulation of the AhR pathway.
Collapse
Affiliation(s)
- Izzah Bungsu
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Nurolaini Kifli
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Siti Rohaiza Ahmad
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Hazim Ghani
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Anne Catherine Cunningham
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| |
Collapse
|
24
|
Chen J, Liu J, Huang Y, Li R, Ma C, Zhang B, Wu F, Yu W, Zuo X, Liang Y, Wang Q. Insights into oral bioavailability enhancement of therapeutic herbal constituents by cytochrome P450 3A inhibition. Drug Metab Rev 2021; 53:491-507. [PMID: 33905669 DOI: 10.1080/03602532.2021.1917598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Herbal plants typically have complex compositions and diverse mechanisms. Among them, bioactive constituents with relatively high exposure in vivo are likely to exhibit therapeutic efficacy. On the other hand, their bioavailability may be influenced by the synergistic effects of different bioactive components. Cytochrome P450 3A (CYP3A) is one of the most abundant CYP enzymes, responsible for the metabolism of 50% of approved drugs. In recent years, many therapeutic herbal constituents have been identified as CYP3A substrates. It is more evident that CYP3A inhibition derived from the herbal formula plays a critical role in improving the oral bioavailability of therapeutic constituents. CYP3A inhibition may be the mechanism of the synergism of herbal formula. In this review, we explored the multiplicity of CYP3A, summarized herbal monomers with CYP3A inhibitory effects, and evaluated herb-mediated CYP3A inhibition, thereby providing new insights into the mechanisms of CYP3A inhibition-mediated oral herb bioavailability.
Collapse
Affiliation(s)
- Junmei Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yueyue Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruoyu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cuiru Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Beiping Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fanchang Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenqian Yu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Zuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
25
|
Identification and characterization of potential antioxidant components in Isodon amethystoides (Benth.) Hara tea leaves by UPLC-LTQ-Orbitrap-MS. Food Chem Toxicol 2021; 148:111961. [DOI: 10.1016/j.fct.2020.111961] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/19/2022]
|
26
|
Fliszár-Nyúl E, Mohos V, Csepregi R, Mladěnka P, Poór M. Inhibitory effects of polyphenols and their colonic metabolites on CYP2D6 enzyme using two different substrates. Biomed Pharmacother 2020; 131:110732. [PMID: 32942157 DOI: 10.1016/j.biopha.2020.110732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Polyphenolic compounds (including flavonoids, chalcones, phenolic acids, and furanocoumarins) represent a common part of our diet, but are also the active ingredients of several dietary supplements and/or medications. These compounds undergo extensive metabolism by human biotransformation enzymes and the microbial flora of the colon. CYP2D6 enzyme metabolizes approximately 25% of the drugs, some of which has narrow therapeutic window. Therefore, its inhibition can lead to the development of pharmacokinetic interactions and the disruption of drug therapy. In this study, the inhibitory effects of 17 plant-derived compounds and 19 colonic flavonoid metabolites on CYP2D6 were examined, employing two assays with different test substrates. The O-demethylation of dextromethorphan was tested employing CypExpress 2D6 kit coupled to HPLC analysis; while the O-demethylation of another CYP2D6 specific substrate (AMMC) was investigated in a plate reader assay with BioVision Fluorometric CYP2D6 kit. Interestingly, some compounds (e.g., bergamottin) inhibited both dextromethorphan and AMMC demethylation; however, certain substances proved to be inhibitors only in one of the assays applied. Our results demonstrate that some polyphenols and colonic metabolites are inhibitors of CYP2D6-catalyzed reactions. Nevertheless, the inhibitory effects showed strong substrate dependence.
Collapse
Affiliation(s)
- Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, H-7624, Pécs, Hungary; Lab-on-a-Chip Research Group, János Szentágothai Research Centre, Ifjúság útja 20, H-7624, Pécs, Hungary.
| | - Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, H-7624, Pécs, Hungary; Lab-on-a-Chip Research Group, János Szentágothai Research Centre, Ifjúság útja 20, H-7624, Pécs, Hungary.
| | - Rita Csepregi
- Lab-on-a-Chip Research Group, János Szentágothai Research Centre, Ifjúság útja 20, H-7624, Pécs, Hungary; Department of Laboratory Medicine, University of Pécs, Medical School, Ifjúság útja 13, H-7624, Pécs, Hungary.
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05, Hradec Králové, Czech Republic.
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, H-7624, Pécs, Hungary; Lab-on-a-Chip Research Group, János Szentágothai Research Centre, Ifjúság útja 20, H-7624, Pécs, Hungary.
| |
Collapse
|
27
|
Inhibitory Effects of Quercetin and Its Main Methyl, Sulfate, and Glucuronic Acid Conjugates on Cytochrome P450 Enzymes, and on OATP, BCRP and MRP2 Transporters. Nutrients 2020; 12:nu12082306. [PMID: 32751996 PMCID: PMC7468908 DOI: 10.3390/nu12082306] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Quercetin is a flavonoid, its glycosides and aglycone are found in significant amounts in several plants and dietary supplements. Because of the high presystemic biotransformation of quercetin, mainly its conjugates appear in circulation. As has been reported in previous studies, quercetin can interact with several proteins of pharmacokinetic importance. However, the interactions of its metabolites with biotransformation enzymes and drug transporters have barely been examined. In this study, the inhibitory effects of quercetin and its most relevant methyl, sulfate, and glucuronide metabolites were tested on cytochrome P450 (CYP) (2C19, 3A4, and 2D6) enzymes as well as on organic anion-transporting polypeptides (OATPs) (OATP1A2, OATP1B1, OATP1B3, and OATP2B1) and ATP (adenosine triphosphate) Binding Cassette (ABC) (BCRP and MRP2) transporters. Quercetin and its metabolites (quercetin-3'-sulfate, quercetin-3-glucuronide, isorhamnetin, and isorhamnetin-3-glucuronide) showed weak inhibitory effects on CYP2C19 and 3A4, while they did not affect CYP2D6 activity. Some of the flavonoids caused weak inhibition of OATP1A2 and MRP2. However, most of the compounds tested proved to be strong inhibitors of OATP1B1, OATP1B3, OATP2B1, and BCRP. Our data demonstrate that not only quercetin but some of its conjugates, can also interact with CYP enzymes and drug transporters. Therefore, high intake of quercetin may interfere with the pharmacokinetics of drugs.
Collapse
|
28
|
Mohos V, Fliszár-Nyúl E, Ungvári O, Bakos É, Kuffa K, Bencsik T, Zsidó BZ, Hetényi C, Telbisz Á, Özvegy-Laczka C, Poór M. Effects of Chrysin and Its Major Conjugated Metabolites Chrysin-7-Sulfate and Chrysin-7-Glucuronide on Cytochrome P450 Enzymes and on OATP, P-gp, BCRP, and MRP2 Transporters. Drug Metab Dispos 2020; 48:1064-1073. [PMID: 32661014 DOI: 10.1124/dmd.120.000085] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022] Open
Abstract
Chrysin is an abundant flavonoid in nature, and it is also contained by several dietary supplements. Chrysin is highly biotransformed in the body, during which conjugated metabolites chrysin-7-sulfate and chrysin-7-glucuronide are formed. These conjugates appear at considerably higher concentrations in the circulation than the parent compound. Based on previous studies, chrysin can interact with biotransformation enzymes and transporters; however, the interactions of its metabolites have been barely examined. In this in vitro study, the effects of chrysin, chrysin-7-sulfate, and chrysin-7-glucuronide on cytochrome P450 enzymes (2C9, 2C19, 3A4, and 2D6) as well as on organic anion-transporting polypeptides (OATPs; 1A2, 1B1, 1B3, and 2B1) and ATP binding cassette [P-glycoprotein, multidrug resistance-associated protein 2, and breast cancer resistance protein (BCRP)] transporters were investigated. Our observations revealed that chrysin conjugates are strong inhibitors of certain biotransformation enzymes (e.g., CYP2C9) and transporters (e.g., OATP1B1, OATP1B3, OATP2B1, and BCRP) examined. Therefore, the simultaneous administration of chrysin-containing dietary supplements with medications needs to be carefully considered due to the possible development of pharmacokinetic interactions. SIGNIFICANCE STATEMENT: Chrysin-7-sulfate and chrysin-7-glucuronide are the major metabolites of flavonoid chrysin. In this study, we examined the effects of chrysin and its conjugates on cytochrome P450 enzymes and on organic anion-transporting polypeptides and ATP binding cassette transporters (P-glycoprotein, breast cancer resistance protein, and multidrug resistance-associated protein 2). Our results demonstrate that chrysin and/or its conjugates can significantly inhibit some of these proteins. Since chrysin is also contained by dietary supplements, high intake of chrysin may interrupt the transport and/or the biotransformation of drugs.
Collapse
Affiliation(s)
- Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Orsolya Ungvári
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Éva Bakos
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Katalin Kuffa
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Tímea Bencsik
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Balázs Zoltán Zsidó
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Csaba Hetényi
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Ágnes Telbisz
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
29
|
Acid Rain Increases Impact of Rice Blast on Crop Health via Inhibition of Resistance Enzymes. PLANTS 2020; 9:plants9070881. [PMID: 32668672 PMCID: PMC7412137 DOI: 10.3390/plants9070881] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023]
Abstract
Worldwide, rice blast (Pyricularia oryzae) causes more rice crop loss than other diseases. Acid rain has reduced crop yields globally for nearly a century. However, the effects of acid rain on rice-Pyricularia oryzae systems are still far from fully understood. In this study, we conducted a lab cultivation experiment of P. oryzae under a series of acidity conditions as well as a glasshouse cultivation experiment of rice that was inoculated with P. oryzae either before (P. + SAR) or after (SAR + P.) simulated acid rain (SAR) at pH 5.0, 4.0, 3.0 and 2.0. Our results showed that the growth and pathogenicity of P. oryzae was significantly inhibited with decreasing pH treatments in vitro culture. The SAR + P. treatment with a pH of 4.0 was associated with the highest inhibition of P. oryzae expansion. However, regardless of the inoculation time, higher-acidity rain treatments showed a decreased inhibition of P. oryzae via disease-resistance related enzymes and metabolites in rice leaves, thus increasing disease index. The combined effects of high acidity and fungal inoculation were more serious than that of either alone. This study provides novel insights into the effects of acid rain on the plant-pathogen interaction and may also serve as a guide for evaluating disease control and crop health in the context of acid rain.
Collapse
|
30
|
Dobiasová S, Řehořová K, Kučerová D, Biedermann D, Káňová K, Petrásková L, Koucká K, Václavíková R, Valentová K, Ruml T, Macek T, Křen V, Viktorová J. Multidrug Resistance Modulation Activity of Silybin Derivatives and Their Anti-inflammatory Potential. Antioxidants (Basel) 2020; 9:antiox9050455. [PMID: 32466263 PMCID: PMC7278776 DOI: 10.3390/antiox9050455] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
Silybin is considered to be the main biologically active component of silymarin. Its oxidized derivative 2,3-dehydrosilybin typically occurs in silymarin in small, but non-negligible amounts (up to 3%). Here, we investigated in detail complex biological activities of silybin and 2,3-dehydrosilybin optical isomers. Antioxidant activities of pure stereomers A and B of silybin and 2,3-dehydrosilybin, as well as their racemic mixtures, were investigated by using oxygen radical absorption capacity (ORAC) and cellular antioxidant activity (CAA) assay. All substances efficiently reduced nitric oxide production and cytokines (TNF-α, IL-6) release in a dose-dependent manner. Multidrug resistance (MDR) modulating potential was evaluated as inhibition of P-glycoprotein (P-gp) ATPase activity and regulation of ATP-binding cassette (ABC) protein expression. All the tested compounds showed strong dose-dependent inhibition of P-gp pump. Moreover, 2,3-dehydrosilybin A (30 µM) displayed the strongest sensitization of doxorubicin-resistant ovarian carcinoma. Despite these significant effects, silybin B was the only compound acting directly upon P-gp in vitro and also downregulating the expression of respective MDR genes. This compound altered the expression of P-glycoprotein (P-gp, ABCB1), multidrug resistance-associated protein 1 (MRP1, ABCC1) and breast cancer resistance protein (BCRP, ABCG2). 2,3-Dehydrosilybin AB exhibited the most effective inhibition of acetylcholinesterase activity. We can clearly postulate that silybin derivatives could serve well as modulators of a cancer drug-resistant phenotype.
Collapse
Affiliation(s)
- Simona Dobiasová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
| | - Kateřina Řehořová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
| | - Denisa Kučerová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
| | - David Biedermann
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic; (D.B.); (L.P.); (K.V.); (V.K.)
| | - Kristýna Káňová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic; (D.B.); (L.P.); (K.V.); (V.K.)
| | - Lucie Petrásková
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic; (D.B.); (L.P.); (K.V.); (V.K.)
| | - Kamila Koucká
- Toxicogenomics Unit, National Institute of Public Health, Šrobárova 49, CZ 100 00 Prague, Czech Republic; (K.K.); (R.V.)
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, CZ 323 00 Pilsen, Czech Republic
| | - Radka Václavíková
- Toxicogenomics Unit, National Institute of Public Health, Šrobárova 49, CZ 100 00 Prague, Czech Republic; (K.K.); (R.V.)
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, CZ 323 00 Pilsen, Czech Republic
| | - Kateřina Valentová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic; (D.B.); (L.P.); (K.V.); (V.K.)
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
| | - Tomáš Macek
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ 142 20 Prague, Czech Republic; (D.B.); (L.P.); (K.V.); (V.K.)
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, CZ 166 28 Prague, Czech Republic; (S.D.); (K.Ř.); (D.K.); (K.K.); (T.R.); (T.M.)
- Correspondence:
| |
Collapse
|
31
|
The Antiproliferative Effects of Flavonoid MAO Inhibitors on Prostate Cancer Cells. Molecules 2020; 25:molecules25092257. [PMID: 32403270 PMCID: PMC7249060 DOI: 10.3390/molecules25092257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 01/23/2023] Open
Abstract
Prostate cancer (PCa) patients commonly experience clinical depression. Recent reports indicated that monoamine oxidase-A (MAO-A) levels elevate in PCa, and antidepressant MAO-Is show anti-PCa properties. In this work, we aimed to find potential drugs for PCa patients suffering from depression by establishing novel anti-PCa reversible monoamine oxidase-A inhibitors (MAO-AIs/RIMA); with an endeavor to understand their mechanism of action. In this investigation, twenty synthesized flavonoid derivatives, defined as KKR compounds were screened for their inhibitory potentials against human MAO-A and MAO-B isozymes. Meanwhile, the cytotoxic and antiproliferative effects were determined in three human PCa cell lines. MAO-A-kinetics, molecular docking, SAR, cell morphology, and cell migration were investigated for the most potent compounds. The screened KKRs inhibited MAO-A more potently than MAO-B, and non-toxically inhibited LNCaP cell proliferation more than the DU145 and PC3 cell lines, respectively. The results showed that the three top MAO-AI KKRs compounds (KKR11, KKR20, and KKR7 (IC50s 0.02-16 μM) overlapped with the top six antiproliferative KKRs against LNCaP (IC50s ~9.4 μM). While KKR21 (MAO-AI) and KKR2A (MAO-I) were ineffective against the PCa cells. Furthermore, KKR21 and KKR11 inhibited MAO-A competitively (Kis ≤ 7.4 nM). Molecular docking of the two compounds predicted shared hydrophobic and distinctive hydrophilic interactions-between the KKR molecule and MAO-A amino acid residues-to be responsible for their reversibility. The combined results and SAR observations indicated that the presence of specific active groups-such as chlorine and hydroxyl groups-are essential in certain MAO-AIs with anti-PCa effects. Additionally, MAO-A inhibition was found to be associated more with anti-PCa property than MAO-B. Distinctively, KKR11 [(E)-3-(3,4-dichlorophenyl)-1-(2-hydroxy-4,6-dimethoxyphenyl)prop-2-en-1-one] exhibited anti-metastatic effects on the DU145 cell line. The chlorine substitution groups might play vital roles in the KKR11 multiple actions. The obtained results indicated that the flavonoid derivative KKR11 could present a novel candidate for PCa patients with depression, through safe non-selective potent inhibition of MAOs.
Collapse
|
32
|
Fang SQ, Huang J, Zhang F, Ni HM, Chen QL, Zhu JR, Fu ZC, Zhu L, Hao WW, Ge GB. Pharmacokinetic interaction between a Chinese herbal formula Huosu Yangwei oral liquid and apatinib in vitro and in vivo. J Pharm Pharmacol 2020; 72:979-989. [PMID: 32285478 DOI: 10.1111/jphp.13268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/14/2020] [Indexed: 12/11/2022]
Abstract
Abstract
Objectives
This study aimed to evaluate the inhibitory effects of Huosu Yangwei oral liquid (HSYW) on cytochrome P450 enzymes (CYPs) and to investigate whether this herbal medicine could modulate the pharmacokinetic behaviour of the co-administered CYP-substrate drug apatinib.
Methods
Cytochrome P450 enzymes inhibition assays were conducted in human liver microsomes (HLM) by a LC-MS/MS method for simultaneous determination of the oxidative metabolites of eight probe substrates for hepatic CYPs. The modulatory effects of HSYW on the oxidative metabolism of apatinib were investigated in both HLM and rat liver microsomes (RLM). The influences of HSYW on the pharmacokinetic behaviour of apatinib were investigated in rats.
Key findings
Huosu Yangwei oral liquid inhibited all tested CYPs in human liver preparations, with the IC50 values ranged from 0.3148 to 2.642 mg/ml. HSYW could also inhibit the formation of two major oxidative metabolites of apatinib in liver microsomes from both human and rat. In-vivo assays demonstrated that HSYW could significantly prolong the plasma half-life of apatinib by 7.4-fold and increase the AUC0–inf (nm·h) of apatinib by 43%, when HSYW (10 ml/kg) was co-administered with apatinib (10 mg/kg) in rats.
Conclusions
Huosu Yangwei oral liquid could inhibit mammalian CYPs and modulated the metabolic half-life of apatinib both in vitro and in vivo.
Collapse
Affiliation(s)
- Sheng-Quan Fang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Huang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Pharmacology and Toxicology Division, Shanghai Institute of Food and Drug Control, Shanghai, China
| | - Feng Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-Mei Ni
- Department of Basic Theory of Traditional Chinese Medicine, College of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi-Long Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun-Ran Zhu
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Chao Fu
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhu
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Qinghai Hospital of Traditional Chinese Medicine, Xining, China
| | - Wei-Wei Hao
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Qinghai Hospital of Traditional Chinese Medicine, Xining, China
| |
Collapse
|
33
|
Park SY, Nguyen PH, Kim G, Jang SN, Lee GH, Phuc NM, Wu Z, Liu KH. Strong and Selective Inhibitory Effects of the Biflavonoid Selamariscina A against CYP2C8 and CYP2C9 Enzyme Activities in Human Liver Microsomes. Pharmaceutics 2020; 12:pharmaceutics12040343. [PMID: 32290339 PMCID: PMC7238120 DOI: 10.3390/pharmaceutics12040343] [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] [Received: 03/25/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Like flavonoids, biflavonoids, dimeric flavonoids, and polyphenolic plant secondary metabolites have antioxidant, antibacterial, antiviral, anti-inflammatory, and anti-cancer properties. However, there is limited data on their effects on cytochrome P450 (P450) and uridine 5'-diphosphoglucuronosyl transferase (UGT) enzyme activities. In this study we evaluate the inhibitory potential of five biflavonoids against nine P450 activities (P450s1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A) in human liver microsomes (HLMs) using cocktail incubation and liquid chromatography-tandem mass spectrometry (LC-MS/MS). The most strongly inhibited P450 activity was CYP2C8-mediated amodiaquine N-dealkylation with IC50 ranges of 0.019~0.123 μM. In addition, the biflavonoids-selamariscina A, amentoflavone, robustaflavone, cupressuflavone, and taiwaniaflavone-noncompetitively inhibited CYP2C8 activity with respective Ki values of 0.018, 0.083, 0.084, 0.103, and 0.142 μM. As selamariscina A showed the strongest effects, we then evaluated it against six UGT isoforms, where it showed weaker inhibition (UGTs1A1, 1A3, 1A4, 1A6, 1A9, and 2B7, IC50 1.7 μM). Returning to the P450 activities, selamariscina A inhibited CYP2C9-mediated diclofenac hydroxylation and tolbutamide hydroxylation with respective Ki values of 0.032 and 0.065 μM in a competitive and noncompetitive manner. However, it only weakly inhibited CYP1A2, CYP2B6, and CYP3A with respective Ki values of 3.1, 7.9, and 4.5 μM. We conclude that selamariscina A has selective and strong inhibitory effects on the CYP2C8 and CYP2C9 isoforms. This information might be useful in predicting herb-drug interaction potential between biflavonoids and co-administered drugs mainly metabolized by CYP2C8 and CYP2C9. In addition, selamariscina A might be used as a strong CYP2C8 and CYP2C9 inhibitor in P450 reaction-phenotyping studies to identify drug-metabolizing enzymes responsible for the metabolism of new chemicals.
Collapse
Affiliation(s)
- So-Young Park
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (G.-H.L.)
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
| | - Phi-Hung Nguyen
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet, Cau Giay, Hanoi 100000, Vietnam;
| | - Gahyun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
| | - Su-Nyeong Jang
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (G.-H.L.)
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
| | - Ga-Hyun Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (G.-H.L.)
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
| | - Nguyen Minh Phuc
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
- Vietnam Hightech of Medicinal and Pharmaceutical JSC, Group 11 Quang Minh town, Hanoi 100000, Vietnam
| | - Zhexue Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (G.-H.L.)
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (G.K.); (N.M.P.); (Z.W.)
- Correspondence: ; Tel.: +82-53-950-8567; Fax: +82-53-950-8557
| |
Collapse
|
34
|
Mohos V, Fliszár-Nyúl E, Lemli B, Zsidó BZ, Hetényi C, Mladěnka P, Horký P, Pour M, Poór M. Testing the Pharmacokinetic Interactions of 24 Colonic Flavonoid Metabolites with Human Serum Albumin and Cytochrome P450 Enzymes. Biomolecules 2020; 10:E409. [PMID: 32155912 PMCID: PMC7175153 DOI: 10.3390/biom10030409] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022] Open
Abstract
Flavonoids are abundant polyphenols in nature. They are extensively biotransformed in enterocytes and hepatocytes, where conjugated (methyl, sulfate, and glucuronide) metabolites are formed. However, bacterial microflora in the human intestines also metabolize flavonoids, resulting in the production of smaller phenolic fragments (e.g., hydroxybenzoic, hydroxyacetic and hydroxycinnamic acids, and hydroxybenzenes). Despite the fact that several colonic metabolites appear in the circulation at high concentrations, we have only limited information regarding their pharmacodynamic effects and pharmacokinetic interactions. Therefore, in this in vitro study, we investigated the interactions of 24 microbial flavonoid metabolites with human serum albumin and cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes. Our results demonstrated that some metabolites (e.g., 2,4-dihydroxyacetophenone, pyrogallol, O-desmethylangolensin, and 2-hydroxy-4-methoxybenzoic acid) form stable complexes with albumin. However, the compounds tested did not considerably displace Site I and II marker drugs from albumin. All CYP isoforms examined were significantly inhibited by O-desmethylangolensin; nevertheless, only its effect on CYP2C9 seems to be relevant. Furthermore, resorcinol and phloroglucinol showed strong inhibitory effects on CYP3A4. Our results demonstrate that, besides flavonoid aglycones and their conjugated derivatives, some colonic metabolites are also able to interact with proteins involved in the pharmacokinetics of drugs.
Collapse
Affiliation(s)
- Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary; (V.M.); (E.F.-N.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; (B.L.)
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary; (V.M.); (E.F.-N.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; (B.L.)
| | - Beáta Lemli
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; (B.L.)
- Institute of Organic and Medicinal Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Balázs Zoltán Zsidó
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary; (B.Z.Z.); (C.H.)
| | - Csaba Hetényi
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary; (B.Z.Z.); (C.H.)
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic; (P.M.)
| | - Pavel Horký
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic; (P.H.)
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Králové, Charles University, Zborovská 2089, 500 05 Hradec Králové, Czech Republic
| | - Milan Pour
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic; (P.H.)
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary; (V.M.); (E.F.-N.)
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; (B.L.)
| |
Collapse
|
35
|
Belitskiy GA, Kirsanov KI, Lesovaya EA, Yakubovskaya MG. Drug-Related Carcinogenesis: Risk Factors and Approaches for Its Prevention. BIOCHEMISTRY (MOSCOW) 2020; 85:S79-S107. [PMID: 32087055 DOI: 10.1134/s0006297920140059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The review summarizes the data on the role of metabolic and repair systems in the mechanisms of therapy-related carcinogenesis and the effect of their polymorphism on the cancer development risk. The carcinogenic activity of different types of drugs, from the anticancer agents to analgesics, antipyretics, immunomodulators, hormones, natural remedies, and non-cancer drugs, is described. Possible approaches for the prevention of drug-related cancer induction at the initiation and promotion stages are discussed.
Collapse
Affiliation(s)
- G A Belitskiy
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia
| | - K I Kirsanov
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia. .,Peoples' Friendship University of Russia, Moscow, 117198, Russia
| | - E A Lesovaya
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia.,Pavlov Ryazan State Medical University, Ryazan, 390026, Russia
| | - M G Yakubovskaya
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
36
|
Burchill MA, Goldberg AR, Tamburini BAJ. Emerging Roles for Lymphatics in Chronic Liver Disease. Front Physiol 2020; 10:1579. [PMID: 31992991 PMCID: PMC6971163 DOI: 10.3389/fphys.2019.01579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic liver disease (CLD) is a global health epidemic causing ∼2 million deaths annually worldwide. As the incidence of CLD is expected to rise over the next decade, understanding the cellular and molecular mediators of CLD is critical for developing novel therapeutics. Common characteristics of CLD include steatosis, inflammation, and cholesterol accumulation in the liver. While the lymphatic system in the liver has largely been overlooked, the liver lymphatics, as in other organs, are thought to play a critical role in maintaining normal hepatic function by assisting in the removal of protein, cholesterol, and immune infiltrate. Lymphatic growth, permeability, and/or hyperplasia in non-liver organs has been demonstrated to be caused by obesity or hypercholesterolemia in humans and animal models. While it is still unclear if changes in permeability occur in liver lymphatics, the lymphatics do expand in number and size in all disease etiologies tested. This is consistent with the lymphatic endothelial cells (LEC) upregulating proliferation specific genes, however, other transcriptional changes occur in liver LECs that are dependent on the inflammatory mediators that are specific to the disease etiology. Whether these changes induce lymphatic dysfunction or if they impact liver function has yet to be directly addressed. Here, we will review what is known about liver lymphatics in health and disease, what can be learned from recent work on the influence of obesity and hypercholesterolemia on the lymphatics in other organs, changes that occur in LECs in the liver during disease and outstanding questions in the field.
Collapse
Affiliation(s)
- Matthew A Burchill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| | - Alyssa R Goldberg
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States.,Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO, United States
| | - Beth A Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| |
Collapse
|
37
|
The Multifarious Link between Cytochrome P450s and Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3028387. [PMID: 31998435 PMCID: PMC6964729 DOI: 10.1155/2020/3028387] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/08/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
Cancer is a leading cause of death worldwide. Cytochrome P450s (P450s) play an important role in the metabolism of endogenous as well as exogenous substances, especially drugs. Moreover, many P450s can serve as targets for disease therapy. Increasing reports of epidemiological, diagnostic, and clinical research indicate that P450s are enzymes that play a major part in the formation of cancer, prevention, and metastasis. The purposes of this review are to shed light on the current state of knowledge about the cancer molecular mechanism involving P450s and to summarize the link between the cancer effects and the participation of P450s.
Collapse
|
38
|
Zhou Y, Li C, Feng B, Chen B, Jin L, Shen Y. UPLC-ESI-MS/MS based identification and antioxidant, antibacterial, cytotoxic activities of aqueous extracts from storey onion (Allium cepa L. var. proliferum Regel). Food Res Int 2019; 130:108969. [PMID: 32156403 DOI: 10.1016/j.foodres.2019.108969] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/16/2019] [Accepted: 12/28/2019] [Indexed: 11/18/2022]
Abstract
Storey onion (Allium cepa L. var. proliferum Regel) is a variety of onion commonly grown in northern China that has not been researched in detail. This study aimed to identify the chemical compositions of storey onion aqueous extracts by UPLC-ESI-MS/MS, as well as characterize the antioxidant, antibacterial and cytotoxic activities, compared with welsh onion and onion. A total of 42 compounds were identified, among which the contents of organosulfur compounds (962.20 ± 34.55 μg/g), polyphenols (100.40 ± 12.55 μg/g) and organic acids (54.04 ± 2.69 μg/g) in storey onion were higher than those in welsh onion and onion. Additionally, the contents of cycloalliin (551.74 ± 8.12 μg/g), ajoene (159.31 ± 5.30 μg/g) and (E)-1-propene-1-sulfenic acid (72.12 ± 2.98 μg/g) in storey onion were the highest. Storey onion had pronounced DPPH• (IC50 = 1.24 ± 0.52 mg/mL) and OH• scavenging activities (IC50 = 14.45 ± 1.29 mg/mL) as well as ferric ion reducing power (absorbance from 0.32 to 2.21). Onion had the highest ABTS•+ scavenging activity (IC50 = 1.64 ± 0.64 mg/mL), while welsh onion had the lowest antioxidant activity. Storey onion had the strongest inhibitory effect on all the tested strains (MIC 31.3-125 mg/mL), and cell viability assays against human liver (HepG2) cancer cell lines also illustrated that aqueous extracts from storey onion significantly inhibited cell proliferation (when incubated for 24 h, IC50 = 33.21 ± 1.12 mg/mL) and induced cell apoptosis. Welsh onion and onion also had weaker antibacterial and anticancer activites, with those of onion being the weakest. The results showed that storey onion with excellent biological activity may benefit to human health and can be developed into functional foods.
Collapse
Affiliation(s)
- Yanyan Zhou
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China
| | - Cong Li
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Bang Feng
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China
| | - Bang Chen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China
| | - Lihua Jin
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China
| | - Yehua Shen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| |
Collapse
|
39
|
Canivenc-Lavier MC, Neiers F, Briand L. Plant polyphenols, chemoreception, taste receptors and taste management. Curr Opin Clin Nutr Metab Care 2019; 22:472-478. [PMID: 31490201 DOI: 10.1097/mco.0000000000000595] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Polyphenols display beneficial health effects through chemopreventive actions on numerous chronic diseases including cancers, metabolic disorders, reproductive disorders and eating behaviour disorders. According to the principle of chemoreception, polyphenols bind cellular targets capable of accepting their stereochemistry, namely metabolizing enzymes and protein receptors, including taste receptors. The extraoral expression of taste receptors and their pharmacological interest in terms of novel drug therapies open up new perspectives on the potential use of these compounds and their interactions with other chemicals in cells. These new perspectives suggest the need to examine these phytochemicals further. However, most polyphenols have a bitterness property that may disrupt the acceptability of healthy foods or dietary supplements. RECENT FINDINGS Polyphenols bind to oral and extraoral bitter type 2 taste receptors, which modulate the signalling pathways involved in anti-inflammatory processes and metabolic and dietary regulations. Depending on their chemical nature, polyphenols may act as activators or inhibitors of taste receptors, and combinations of polyphenols (or herbal mixtures) may be used to modulate the acceptability of bitterness. SUMMARY The current review summarizes recent findings on polyphenol chemoreception and highlights the evidence of healthy effects through type 2 taste receptor mediation in signalling pathways, such as new targets, with promising perspectives.
Collapse
Affiliation(s)
- Marie-Chantal Canivenc-Lavier
- Centre des Sciences du GoÛt et de l'Alimentation (CSGA), INRA, Université de Bourgogne Franche-Comté, AgroSup, CNRS, Dijon, France
| | | | | |
Collapse
|
40
|
Interactions of 7,8-Dihydroxyflavone with Serum Albumin as well as with CYP2C9, CYP2C19, CYP3A4, and Xanthine Oxidase Biotransformation Enzymes. Biomolecules 2019; 9:biom9110655. [PMID: 31731555 PMCID: PMC6920897 DOI: 10.3390/biom9110655] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023] Open
Abstract
7,8-dihydroxyflavone (DHF) is a flavone aglycone which has beneficial effects in several central nervous system diseases. Most of the pharmacokinetic properties of DHF have been characterized, while only limited information is available regarding its interactions with serum albumin and biotransformation enzymes. In this study, the interactions of DHF with albumin was examined employing fluorescence spectroscopy and ultrafiltration. Furthermore, the inhibitory effects of DHF on cytochrome P450 (CYP2C9, CYP2C19, and CYP3A4) and xanthine oxidase (XO) enzymes were also tested using in vitro models. Our results demonstrate that DHF forms a stable complex with albumin (K = 4.9 × 105 L/mol) and that it is able to displace both Site I and Site II ligands. Moreover, DHF proved to be a potent inhibitor of each enzyme tested, showing similar or slightly weaker effects than the positive controls used. Considering the above-listed observations, the coadministration of DHF with drugs may interfere with the drug therapy due to the development of pharmacokinetic interactions.
Collapse
|
41
|
Hernáez Á, Estruch R. The Mediterranean Diet and Cancer: What Do Human and Molecular Studies Have to Say about It? Nutrients 2019; 11:E2155. [PMID: 31505794 PMCID: PMC6769497 DOI: 10.3390/nu11092155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 02/07/2023] Open
Abstract
Mediterranean diet (MD) is a well-known healthy dietary pattern, linked to: (1) high intakes of olive oil as main the culinary fat, plant-based foods (fruits, vegetables, legumes, whole grains, tree nuts, and seeds), and fish; and (2) a moderate consumption of white meat, eggs, dairy products such as yogurt and cheese, and wine always with meals [...].
Collapse
Affiliation(s)
- Álvaro Hernáez
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.
- CIBER of Pathophysiology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Ramón Estruch
- CIBER of Pathophysiology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Internal Medicine, Hospital Clínic, University of Barcelona, 08036 Barcelona, Spain.
| |
Collapse
|
42
|
Modulation of hepatic ABC transporters by Eruca vesicaria intake: Potential diet-drug interactions. Food Chem Toxicol 2019; 133:110797. [PMID: 31479713 DOI: 10.1016/j.fct.2019.110797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/02/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023]
Abstract
The aim of this work was to evaluate whether oral administration of Eruca vesicaria, a species of rocket cultivated in Argentina, could modify cyclophosphamide (CP)-induced genotoxicity through modulation of hepatic ABC transporters. Daily oral administration of E. vesicaria fresh leaves juice (1.0, 1.4 and 2.0 g/kg) for 14 days did not alter genotoxicity biomarkers -alkaline comet assay and micronucleus test -in neither male nor female mice. Instead, repeated intake of this cruciferous decreased CP-induced DNA damage dose-dependently and it caused hepatic overexpression of P-glycoprotein (P-gp; 1.4 and 2.0 g/kg) and multidrug resistance protein 2 (MRP2; 2.0 g/kg), but not breast cancer resistance protein (Bcrp). The antigenotoxic effect of E. vesicaria was prevented by 50 mg/kg verapamil (P-gp inhibitor) or 10 mg/kg indomethacin (MRP2 inhibitor). In turn, CP-induced cytotoxicity (10 mM, 24 h) on human hepatoma cells (HepG2/C3A) was significantly reduced by preincubation with E. vesicaria (1.4 mg/ml; 48 h); this effect was absent when CP was coincubated with 35 μM verapamil, 80 μM indomethacin or 10 μM KO-143 (BCRP inhibitor). Altogether, these results allow us to demonstrate that repeated intake of E. vesicaria exhibited antigenotoxicity, at least in part, by induction of hepatic ABC transporters in vivo in mice as well as in vitro in human liver cells. This could account for other diet-drug interactions.
Collapse
|
43
|
Vida RG, Fittler A, Somogyi-Végh A, Poór M. Dietary quercetin supplements: Assessment of online product informations and quantitation of quercetin in the products by high-performance liquid chromatography. Phytother Res 2019; 33:1912-1920. [PMID: 31155780 DOI: 10.1002/ptr.6382] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/31/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
Administration of the increasingly popular dietary supplements containing quercetin may interfere with drug therapy. We intended to evaluate the online availability and quercetin content of the high-dose mono-component quercetin products and to review the potential use of quercetin products and their interactions with drugs. We monitored the online access to quercetin-containing dietary supplements, collected the relevant information from the websites, procured selected products from the vendors, and subjected them to substance analysis. The quercetin content was quantified by an HPLC-UV method. Twenty-five websites offered mono-component quercetin products, and nine products were procured. The quercetin content of eight products differed only ±10% from the nominal dose, whereas one product contained almost 30% more quercetin. Misleading indications such as antitumor and cardiovascular effects were often found on the sellers' websites. Quercetin-containing dietary supplements are available online with misleading indications. The recommended daily doses are often high (occasionally over 1,000 mg), which may induce clinically relevant interactions with medications. Because high-quercetin content of dietary supplements was confirmed, health care professionals should be aware of the unregulated internet market of dietary supplements and should consider the interactions of these substances with drugs.
Collapse
Affiliation(s)
- Róbert György Vida
- Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - András Fittler
- Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Anna Somogyi-Végh
- Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Miklós Poór
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| |
Collapse
|
44
|
Inhibitory Effects of Quercetin and Its Human and Microbial Metabolites on Xanthine Oxidase Enzyme. Int J Mol Sci 2019; 20:ijms20112681. [PMID: 31159151 PMCID: PMC6600370 DOI: 10.3390/ijms20112681] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Quercetin is an abundant flavonoid in nature and is used in several dietary supplements. Although quercetin is extensively metabolized by human enzymes and the colonic microflora, we have only few data regarding the pharmacokinetic interactions of its metabolites. Therefore, we investigated the interaction of human and microbial metabolites of quercetin with the xanthine oxidase enzyme. Inhibitory effects of five conjugates and 23 microbial metabolites were examined with 6-mercaptopurine and xanthine substrates (both at 5 μM), employing allopurinol as a positive control. Quercetin-3'-sulfate, isorhamnetin, tamarixetin, and pyrogallol proved to be strong inhibitors of xanthine oxidase. Sulfate and methyl conjugates were similarly strong inhibitors of both 6-mercaptopurine and xanthine oxidations (IC50 = 0.2-0.7 μM); however, pyrogallol inhibited xanthine oxidation (IC50 = 1.8 μM) with higher potency vs. 6-MP oxidation (IC50 = 10.1 μM). Sulfate and methyl conjugates were approximately ten-fold stronger inhibitors (IC50 = 0.2-0.6 μM) of 6-mercaptopurine oxidation than allopurinol (IC50 = 7.0 μM), and induced more potent inhibition compared to quercetin (IC50 = 1.4 μM). These observations highlight that some quercetin metabolites can exert similar or even a stronger inhibitory effect on xanthine oxidase than the parent compound, which may lead to the development of quercetin-drug interactions (e.g., with 6-mercaptopurin or azathioprine).
Collapse
|
45
|
The Flavonoid Quercetin Induces AP-1 Activation in FRTL-5 Thyroid Cells. Antioxidants (Basel) 2019; 8:antiox8050112. [PMID: 31035637 PMCID: PMC6562732 DOI: 10.3390/antiox8050112] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022] Open
Abstract
Previous studies have shown that quercetin inhibits thyroid function both in vitro and in vivo. An attempt to evaluate the effect of quercetin at the promoter level of the thyroid-specific genes led to the observation that this compound induces the basal activity of the reporter vector. Therefore, the action of quercetin has been evaluated on the basal activity of several reporter vectors: The PGL3 basic, promoter and control vectors from Promega, and a pSV-based chloramphenicol acetyltransferase (CAT) reporter vector. In the Fisher Rat Thyroid cell Line FRTL-5 thyroid cells transiently transfected, quercetin 10 μM increased the basal activity of all the reporter vectors evaluated, although the degree of the effect was significantly different among them. The analysis of the difference among the regulatory regions of these vectors identified the activator protein 1 (AP-1) binding site as one of the potential sites involved in the quercetin effect. Electromobility shift assay experiments showed that the treatment with quercetin induced the binding of a protein complex to an oligonucleotide containing the AP-1 consensus binding site. This is the first study showing an effect of quercetin on AP-1 activity in thyroid cells. Further studies are in progress to understand the role of AP-1 activation in the effects of quercetin on thyroid function.
Collapse
|
46
|
Ghadiri S, Spalenza V, Dellafiora L, Badino P, Barbarossa A, Dall'Asta C, Nebbia C, Girolami F. Modulation of aflatoxin B1 cytotoxicity and aflatoxin M1 synthesis by natural antioxidants in a bovine mammary epithelial cell line. Toxicol In Vitro 2019; 57:174-183. [PMID: 30849473 DOI: 10.1016/j.tiv.2019.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/14/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Abstract
Aflatoxin (AF) B1, a widespread food and feed contaminant, is bioactivated by drug metabolizing enzymes (DME) to cytotoxic and carcinogenic metabolites like AFB1-epoxide and AFM1, a dairy milk contaminant. A number of natural antioxidants have been reported to afford a certain degree of protection against AFB1 (cyto)toxicity. As the mammary gland potentially participates in the generation of AFB1 metabolites, we evaluated the role of selected natural antioxidants (i.e. curcumin, quercetin and resveratrol) in the modulation of AFB1 toxicity and metabolism using a bovine mammary epithelial cell line (BME-UV1). Quercetin and, to a lesser extent, resveratrol and curcumin from Curcuma longa (all at 5 μM) significantly counteracted the AFB1-mediated impairment of cell viability (concentration range: 96-750 nM). Moreover, quercetin was able to significantly reduce the synthesis of AFM1. The quantitative PCR analysis on genes encoding for DME (phase I and II) and antioxidant enzymes showed that AFB1 caused an overall downregulation of the detoxifying systems, and mainly of GSTA1, which mediates the GSH conjugation of the AFB1-epoxide. The negative modulation of GSTA1 was efficiently reversed in the presence of quercetin, which significantly increased GSH levels as well. It is suggested that quercetin exerts its beneficial effects by depressing the bio-transformation of AFB1 and counterbalancing its pro-oxidant effects.
Collapse
Affiliation(s)
- Shiva Ghadiri
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, Grugliasco, Italy
| | - Veronica Spalenza
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, Grugliasco, Italy
| | - Luca Dellafiora
- Department of Food and Drug, University of Parma, Via G.P. Usberti 27/A, 43124 Parma, Italy
| | - Paola Badino
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, Grugliasco, Italy
| | - Andrea Barbarossa
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, Ozzano dell'Emilia, Italy
| | - Chiara Dall'Asta
- Department of Food and Drug, University of Parma, Via G.P. Usberti 27/A, 43124 Parma, Italy
| | - Carlo Nebbia
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, Grugliasco, Italy
| | - Flavia Girolami
- Department of Veterinary Sciences, University of Torino, Largo Braccini 2, Grugliasco, Italy.
| |
Collapse
|
47
|
Rasouli H, Hosseini-Ghazvini SMB, Khodarahmi R. Therapeutic Potentials of the Most Studied Flavonoids: Highlighting Antibacterial and Antidiabetic Functionalities. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2019. [DOI: 10.1016/b978-0-444-64181-6.00003-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
48
|
Yu C, Chen DQ, Liu HX, Li WB, Lu JW, Feng JF. Rosmarinic acid reduces the resistance of gastric carcinoma cells to 5-fluorouracil by downregulating FOXO4-targeting miR-6785-5p. Biomed Pharmacother 2019; 109:2327-2334. [PMID: 30551491 DOI: 10.1016/j.biopha.2018.10.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Chemoresistance has been a major problem in cancer chemotherapy. The present study aimed to investigate the effect of Rosmarinic acid (RA) on chemoresistance to 5-Fu and its molecular mechanism in gastric carcinoma. METHODS CCK8 cell proliferation and apoptosis assay were used to evaluate the effect of RA on chemoresistance to 5-Fu in GC cells. RNA microarray was used to identify miRNA involved. Expression level of miRNA in GC cells was determined by RT-PCR. Down- or up-regulating of miRNA in the GC cells was performed by transfection of RNA interference or expression vectors in the GC cells. Double luciferase reporter assay was used to verify miRNA target genes. Expression of P-glycoprotein and Bax was analyzed with Western blot. RESULTS RA treated SGC7901/5-Fu cells showed significant increased chemosensitivity to 5-Fu. The IC50 of 5-Fu was significantly reduced in RA treated SGC7901/5-Fu cells (70.43 ± 1.06 μg/mL) compared to untreated SGC7901/5-Fu cells (208.6 ± 1.09 μg/mL) (P < 0.05). Apoptosis rate was significantly increased in RA+5-Fu treated SGC7901/5-Fu cells compared to 5-FU treatment alone (P < 0.01). Two miRNAs, namely miR-642a-3p and miR-6785-5p, were identified to be involved in the chemo-sensitizing effect of RA in the SGC7901/5-Fu cells. RA treated SGC7901/5-Fu cells showed reduced expression levels of miR-642a-3p and miR-6785-5p compared to untreated SGC7901/5-Fu cells (P < 0.05). Down- or up-regulation of miR-6785-5p increased or reduced chemosensitivity of gastric carcinoma cells to 5-Fu, respectively. RA treated SGC7901/5-Fu and the SGC7901/5-Fu-Si cells showed significantly increased FOXO4 expression (P < 0.01). Double luciferase reporter assay confirmed miR-6785-5p directly targets FOXO4 to regulate its expression. RA significantly reduced P-gp expression and increased Bax expression in SGC7901/5-Fu and the SGC7901/5-Fu-Si cells (P < 0.05). CONCLUSION RA enhances chemosensitivity of resistant gastric carcinoma SGC7901 cells to 5-Fu by downregulating miR-6785-5p and miR-642a-3p and increasing FOXO4 expression. These study suggest the potential for RA as a multidrug resistance-reversing agent in GC.
Collapse
Affiliation(s)
- Chen Yu
- Department of Integrated TCM & Western Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiang Su, 210000
| | - Dong-Qing Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nan Jing Medical University Affiliated Cancer Hospital, Suzhou, Jiang Su, 210000
| | - Hai-Xia Liu
- Department of Oncology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiang Su, 210000
| | - Wei-Bing Li
- Department of Integrated TCM & Western Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiang Su, 210000
| | - Jian-Wei Lu
- Department of Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiang Su, 210000.
| | - Ji-Feng Feng
- Department of Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiang Su, 210000.
| |
Collapse
|
49
|
Liu XY, Lv X, Wang P, Ai CZ, Zhou QH, Finel M, Fan B, Cao YF, Tang H, Ge GB. Inhibition of UGT1A1 by natural and synthetic flavonoids. Int J Biol Macromol 2018; 126:653-661. [PMID: 30594625 DOI: 10.1016/j.ijbiomac.2018.12.171] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 01/27/2023]
Abstract
Flavonoids are widely distributed phytochemicals in vegetables, fruits and medicinal plants. Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. In this study, the inhibitory effects of 56 natural and synthetic flavonoids on UGT1A1 were assayed, while the structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated. The results demonstrated that the C-3 and C-7 hydroxyl groups on the flavone skeleton would enhance UGT1A1 inhibition, while flavonoid glycosides displayed weaker inhibitory effects than their corresponding aglycones. Further investigation on inhibition kinetics of two strong flavonoid-type UGT1A1 inhibitors, acacetin and kaempferol, yielded interesting results. Both flavonoids were competitive inhibitors against UGT1A1-mediated NHPN-O-glucuronidation, but were mixed and competitive inhibitors toward UGT1A1-mediated NCHN-O-glucuronidation, respectively. Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition.
Collapse
Affiliation(s)
- Xin-Yu Liu
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 200473, China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang 832000, China
| | - Xia Lv
- Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, China
| | - Ping Wang
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 200473, China
| | - Chun-Zhi Ai
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Qi-Hang Zhou
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 200473, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014, Finland
| | - Bin Fan
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 200473, China
| | - Yun-Feng Cao
- Key Laboratory of Contraceptives and Devices Research (NPFPC), Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute of Planned Parenthood Research, Shanghai 200032, China
| | - Hui Tang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang 832000, China.
| | - Guang-Bo Ge
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 200473, China.
| |
Collapse
|
50
|
Mohos V, Fliszár-Nyúl E, Schilli G, Hetényi C, Lemli B, Kunsági-Máté S, Bognár B, Poór M. Interaction of Chrysin and Its Main Conjugated Metabolites Chrysin-7-Sulfate and Chrysin-7-Glucuronide with Serum Albumin. Int J Mol Sci 2018; 19:ijms19124073. [PMID: 30562928 PMCID: PMC6320863 DOI: 10.3390/ijms19124073] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022] Open
Abstract
Chrysin (5,7-dihydroxyflavone) is a flavonoid aglycone, which is found in nature and in several dietary supplements. During the biotransformation of chrysin, its conjugated metabolites chrysin-7-sulfate (C7S) and chrysin-7-glucuronide (C7G) are formed. Despite the fact that these conjugates appear in the circulation at much higher concentrations than chrysin, their interactions with serum albumin have not been reported. In this study, the complex formation of chrysin, C7S, and C7G with human (HSA) and bovine (BSA) serum albumins was investigated employing fluorescence spectroscopic, ultrafiltration, and modeling studies. Our major observations/conclusions are as follows: (1) Compared to chrysin, C7S binds with a threefold higher affinity to HSA, while C7G binds with a threefold lower affinity; (2) the albumin-binding of chrysin, C7S, and C7G did not show any large species differences regarding HSA and BSA; (3) tested flavonoids likely occupy Sudlow’s Site I in HSA; (4) C7S causes significant displacement of Sudlow’s Site I ligands, exerting an even stronger displacing ability than the parent compound chrysin. Considering the above-listed observations, the high intake of chrysin (e.g., through the consumption of dietary supplements with high chrysin contents) may interfere with the albumin-binding of several drugs, mainly due to the strong interaction of C7S with HSA.
Collapse
Affiliation(s)
- Violetta Mohos
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Szigeti út 12, H-7624 Pécs, Hungary.
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary.
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Szigeti út 12, H-7624 Pécs, Hungary.
| | - Gabriella Schilli
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, Szigeti út 12, H-7624 Pécs, Hungary.
| | - Csaba Hetényi
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, Szigeti út 12, H-7624 Pécs, Hungary.
| | - Beáta Lemli
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary.
- Department of Pharmaceutical Chemistry, University of Pécs, Faculty of Pharmacy, Rókus utca 2, H-7624 Pécs, Hungary.
| | - Sándor Kunsági-Máté
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary.
- Department of Pharmaceutical Chemistry, University of Pécs, Faculty of Pharmacy, Rókus utca 2, H-7624 Pécs, Hungary.
| | - Balázs Bognár
- Department of Organic and Pharmacological Chemistry, University of Pécs, Medical School, Honvéd utca 1, H-7624 Pécs, Hungary.
| | - Miklós Poór
- Department of Pharmacology, University of Pécs, Faculty of Pharmacy, Szigeti út 12, H-7624 Pécs, Hungary.
- János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary.
| |
Collapse
|