1
|
Li M, Wang Y, Chen Y, Dong L, Liu J, Dong Y, Yang Q, Cai W, Li Q, Peng B, Li Y, Weng X, Wang Y, Zhu X, Gong Z, Chen Y. A comprehensive review on pharmacokinetic mechanism of herb-herb/drug interactions in Chinese herbal formula. Pharmacol Ther 2024; 264:108728. [PMID: 39389315 DOI: 10.1016/j.pharmthera.2024.108728] [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: 02/28/2024] [Revised: 07/16/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
Oral administration of Chinese Herbal Medicine (CHM) faces various challenges in reaching the target organs including absorption and conversion in the gastrointestinal tract, hepatic metabolism via the portal vein, and eventual systemic circulation. During this process, factors such as gut microbes, physical or chemical barriers, metabolic enzymes, and transporters play crucial roles. Particularly, interactions between different herbs in CHM have been observed both in vitro and in vivo. In vitro, interactions typically manifest as detectable physical or chemical changes, such as facilitating solubilization or producing precipitates when decoctions of multiple herbs are administered. In vivo, such interactions cause alterations in the ADME (absorption, distribution, metabolism, and excretion) profile on metabolic enzymes or transporters in the body, leading to competition, antagonism, inhibition, or activation. These interactions ultimately contribute to differences in the therapeutic and pharmacological effects of multi-herb formulas in CHM. Over the past two thousand years, China has cultivated profound expertise and solid theoretical frameworks over the scientific use of herbs. The combination of multiple herbs in one decoction has been frequently employed to synergistically enhance therapeutic efficacy or mitigate toxic and side effects in clinical settings. Additionally combining herbs with increased toxicity or decreased effect is also regarded as a remedy, a practice that should be approached with caution according to Traditional Chinese Medicine (TCM) physicians. Such historical records and practices serve as a foundation for predicting favorable multi-herb combinations and their potential risks. However, systematic data that are available to support the clinical practice and the exploration of novel herbal formulas remain limited. Therefore, this review aims to summarize the pharmacokinetic interactions and mechanisms of herb-herb or herb-drug combinations from existing works, and to offer guidance as well as evidence for optimizing CHM and developing new medicines with CHM characteristics.
Collapse
Affiliation(s)
- Mengting Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yanli Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Lijinchuan Dong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jieyuan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yu Dong
- Guang'an men hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Weiyan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bo Peng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yajie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Zipeng Gong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China.
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
2
|
Huang L, Luo S, Tong S, Lv Z, Wu J. The development of nanocarriers for natural products. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1967. [PMID: 38757428 DOI: 10.1002/wnan.1967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/01/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
Natural bioactive compounds from plants exhibit substantial pharmacological potency and therapeutic value. However, the development of most plant bioactive compounds is hindered by low solubility and instability. Conventional pharmaceutical forms, such as tablets and capsules, only partially overcome these limitations, restricting their efficacy. With the recent development of nanotechnology, nanocarriers can enhance the bioavailability, stability, and precise intracellular transport of plant bioactive compounds. Researchers are increasingly integrating nanocarrier-based drug delivery systems (NDDS) into the development of natural plant compounds with significant success. Moreover, natural products benefit from nanotechnological enhancement and contribute to the innovation and optimization of nanocarriers via self-assembly, grafting modifications, and biomimetic designs. This review aims to elucidate the collaborative and reciprocal advancement achieved by integrating nanocarriers with botanical products, such as bioactive compounds, polysaccharides, proteins, and extracellular vesicles. This review underscores the salient challenges in nanomedicine, encompassing long-term safety evaluations of nanomedicine formulations, precise targeting mechanisms, biodistribution complexities, and hurdles in clinical translation. Further, this study provides new perspectives to leverage nanotechnology in promoting the development and optimization of natural plant products for nanomedical applications and guiding the progression of NDDS toward enhanced efficiency, precision, and safety. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Liying Huang
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Shicui Luo
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Sen Tong
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhuo Lv
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Junzi Wu
- The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Clinical Medical Research Center for Geriatric Diseases, Yunnan First People's Hospital, Kunming, Yunnan, China
| |
Collapse
|
3
|
Ding C, Guo C, Fang L, Li Y, Wang Z, Dong Z. Determination of vericiguat in rat plasma by UPLC-MS/MS and its application to drug interaction. J Chromatogr A 2023; 1709:464401. [PMID: 37741219 DOI: 10.1016/j.chroma.2023.464401] [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] [Received: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Vericiguat (VER) is a novel soluble guanylate cyclase stimulator treating symptomatic chronic heart failure (HF), and it is a substrate of both transporters P-glycoprotein and breast cancer resistance protein (BCRP). Astragaloside IV (ASIV) is the main active ingredient in Radix Astragali (Huangqi), a traditional Chinese medicine widely used for HF treatment in China. ASIV's effect on the protein expression of P-glycoprotein and BCRP has been observed, its impact on VER metabolism remain uncertain. In the present study, male Sprague-Dawley rats were administered with 20 mg/kg ASIV and 1 mg/kg VER to study their pharmacokinetics. Blood samples were subject to liquid-liquid extraction, and riociguat was employed as the internal standard (IS). The analytical method involved a C18 column (XSelect® HSS T3 column, 2.1 × 100 mm, 2.5 μm) with a mobile phase of 0.1% formic acid and acetonitrile for gradient elution. The flow rate of the mobile phase was set at 0.2 mL/min, and 5 µL of the sample was used for analysis. The positive ion multi-response monitoring mode was utilized with a transition of m/z 427.4→109.1 for VER and m/z 423.3→109.1 for the IS. The method exhibited good linearity within the concentration range of 0.1 to 300 ng/mL (r = 0.9987), and all the validation processes were conducted in accordance with the requirements of biological analysis. The pharmacokinetic results revealed that ASIV did not significantly alter the main parameters of VER, except for Cmax, which decreased by 33.2% (P < 0.05). Overall, our study successfully established a selective, sensitive and repeatable ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) analysis for detecting VER in rat plasma.
Collapse
Affiliation(s)
- Congyang Ding
- National Clinical Drug Monitoring Center, Hebei General Hospital, Shijiazhuang, Hebe, 050051, China
| | - Caihui Guo
- National Clinical Drug Monitoring Center, Hebei General Hospital, Shijiazhuang, Hebe, 050051, China
| | - Lingzhi Fang
- National Clinical Drug Monitoring Center, Hebei General Hospital, Shijiazhuang, Hebe, 050051, China
| | - Yajing Li
- National Clinical Drug Monitoring Center, Hebei General Hospital, Shijiazhuang, Hebe, 050051, China
| | - Zhi Wang
- Graduate school, Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Zhanjun Dong
- National Clinical Drug Monitoring Center, Hebei General Hospital, Shijiazhuang, Hebe, 050051, China; Graduate school, Hebei Medical University, Shijiazhuang, Hebei 050051, China.
| |
Collapse
|
4
|
Melin LG, Dall JH, Lindholt JS, Steffensen LB, Beck HC, Elkrog SL, Clausen PD, Rasmussen LM, Stubbe J. Cycloastragenol Inhibits Experimental Abdominal Aortic Aneurysm Progression. Biomedicines 2022; 10:biomedicines10020359. [PMID: 35203568 PMCID: PMC8962318 DOI: 10.3390/biomedicines10020359] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm involves vascular inflammation and elastin degradation. Astragalusradix contains cycloastragenol, which is known to be anti-inflammatory and to protect against elastin degradation. We hypothesized that cycloastragenol supplementation inhibits abdominal aortic aneurysm progression. Abdominal aortic aneurysm was induced in male rats by intraluminal elastase infusion in the infrarenal aorta and treated daily with cycloastragenol (125 mg/kg/day). Aortic expansion was followed weekly by ultrasound for 28 days. Changes in aneurysmal wall composition were analyzed by mRNA levels, histology, zymography and explorative proteomic analyses. At day 28, mean aneurysm diameter was 37% lower in the cycloastragenol group (p < 0.0001). In aneurysm cross sections, elastin content was insignificantly higher in the cycloastragenol group (10.5% ± 5.9% vs. 19.9% ± 16.8%, p = 0.20), with more preserved elastin lamellae structures (p = 0.0003) and without microcalcifications. Aneurysmal matrix metalloprotease-2 activity was reduced by the treatment (p = 0.022). Messenger RNA levels of inflammatory- and anti-oxidative markers did not differ between groups. Explorative proteomic analysis showed no difference in protein levels when adjusting for multiple testing. Among proteins displaying nominal regulation were fibulin-5 (p = 0.02), aquaporin-1 (p = 0.02) and prostacyclin synthase (p = 0.007). Cycloastragenol inhibits experimental abdominal aortic aneurysm progression. The suggested underlying mechanisms involve decreased matrix metalloprotease-2 activity and preservation of elastin and reduced calcification, thus, cycloastragenol could be considered for trial in abdominal aortic aneurysm patients.
Collapse
Affiliation(s)
- Leander Gaarde Melin
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Julie Husted Dall
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Jes S. Lindholt
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lasse B. Steffensen
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Hans Christian Beck
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Sophie L. Elkrog
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Pernille D. Clausen
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Jane Stubbe
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
- Correspondence: ; Tel.: +45-6550-3709
| |
Collapse
|
5
|
Liu JX, Zheng XY, Zhang YH, Song WT, Chang D. Research progress on the pharmacological mechanisms of chinese medicines that tonify Qi and activate blood against cerebral ischemia/reperfusion injury. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2022. [DOI: 10.4103/wjtcm.wjtcm_21_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
6
|
Kumar S, Bouic PJ, Rosenkranz B. Investigation of CYP2B6, 3A4 and β-esterase interactions of Withania somnifera (L.) dunal in human liver microsomes and HepG2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113766. [PMID: 33395575 DOI: 10.1016/j.jep.2020.113766] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 10/27/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Withania somnifera (L.) Dunal (Solanaceae) is a traditional herb, used in African indigenous systems of medicine for the treatment of various diseases (including HIV/AIDS and tuberculosis). The relevance of clinically significant interactions of Withania with ARVs and anti-TB drugs needs to be investigated. AIM OF THE STUDY This study evaluated the effects of its roots on cytochromes P450 (CYPs) 2B6, 3A4, and rifampicin metabolism pathway, using methanol, ethanol, aqueous, and ethyl acetate solvent extractions. MATERIALS AND METHODS The extracts were tested on human liver microsomes (HLM) for CYP inhibition, mRNA expression in HepG2 cells for CYP induction. Biochemical qualitative tests and LC-MS/MS methodology were used to determine active phytoconstituents. RESULTS The methanolic and ethyl acetate extracts inhibited CYP2B6 with IC50s 79.16 and 57.96 μg/ml respectively, while none of the extracts had any effect on rifampicin metabolism or showed time-dependant inhibition (TDI). All extracts were moderate inducers of CYP3A4; the aqueous extract exhibited 38%-fold shift induction of CYP3A4 compared to the control. The methanolic extract had the lowest CTC50 (50% of cytotoxicity inhibition) (67.13 ± 0.83 μg/ml). LC-MS/MS-PDA full scans were consistent with the presence of flavone salvigenin (m/z 327), alkaloid isopelletierine (m/z 133), steroidal lactone 2,3-dihydrowithaferin-A (m/z 472), and other withanolides including withaperuvin I (m/z 533), withaferin derivative (m/z 567), some of these compounds likely being responsible for the observed CYP2B6 inhibition and CYP3A4 induction. The putative gastrointestinal tract (GIT) concentration for the active extracts was 1800 μg/ml and the hepatic circulation concentrations were estimated at about 220 μg/ml and 13.5 μg/ml for the methanolic and ethyl acetate extracts, respectively. The extrapolated in vivo percentage of inhibition was at 85% for the methanolic extract against CYP2B6. CONCLUSIONS The findings reported in this study suggest that W. somnifera extracts have the potential of causing clinically significant herb-drug interactions (HDI) as moderate inducer of CYP3A4 and inhibitor of CYP2B6 metabolism pathway (methanol and ethyl acetate extracts).
Collapse
Affiliation(s)
- Saneesh Kumar
- Division of Clinical Pharmacology, University of Stellenbosch, Cape Town, South Africa.
| | - Patrick J Bouic
- Division of Medical Microbiology, University of Stellenbosch, Cape Town, South Africa; Synexa Life Sciences, Montague Gardens, Cape Town, South Africa.
| | - Bernd Rosenkranz
- Division of Clinical Pharmacology, University of Stellenbosch, Cape Town, South Africa.
| |
Collapse
|
7
|
Wu J, Zeng Z, Li Y, Qin H, Zuo C, Zhou C, Xu D. Cycloastragenol protects against glucocorticoid-induced osteogenic differentiation inhibition by activating telomerase. Phytother Res 2020; 35:2034-2044. [PMID: 33165990 DOI: 10.1002/ptr.6946] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/17/2020] [Accepted: 10/25/2020] [Indexed: 12/25/2022]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) that is mainly featured as low bone density and increased risk of fracture is prone to occur with the administration of excessive glucocorticoids. Cycloastragenol (CAG) has been verified to be a small molecule that activates telomerase. Studied showed that up-regulated telomerase was associated with promoting osteogeneic differentiation, so we explored whether CAG could promote osteogenic differentiation to protect against GIOP and telomerase would be the target that CAG exerted its function. Our results demonstrated that CAG prominently increased the ALP activity, mineralization, mRNA of runt-related transcription factor 2, osteocalcin, osteopontin, collagen type I in both MC3T3-E1 cells and dexamethasone (DEX)-treated MC3T3-E1 cells. CAG up-regulated telomerase reverse transcriptase and the protective effect of CAG was blocked by telomerase inhibitor TMPyP4. Moreover, CAG improved bone mineralization in DEX-induced bone damage in a zebrafish larvea model. Therefore, the study showed that CAG could alleviate the osteogenic differentiation inhibition induced by DEX in vitro and in vivo, and CAG might be considered as a candidate drug for the treatment of GIOP.
Collapse
Affiliation(s)
- Jiahuan Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Zhanwei Zeng
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Yuyun Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Huiyi Qin
- Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Changqing Zuo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Chenhui Zhou
- School of Nursing, Guangdong Medical University, Dongguan, China
| | - Daohua Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| |
Collapse
|
8
|
Qin Z, Jia M, Yang J, Xing H, Yin Z, Yao Z, Zhang X, Yao X. Multiple circulating alkaloids and saponins from intravenous Kang-Ai injection inhibit human cytochrome P450 and UDP-glucuronosyltransferase isozymes: potential drug-drug interactions. Chin Med 2020; 15:69. [PMID: 32655683 PMCID: PMC7339578 DOI: 10.1186/s13020-020-00349-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Kang-Ai injection is widely used as an adjuvant therapy drug for many cancers, leukopenia, and chronic hepatitis B. Circulating alkaloids and saponins are believed to be responsible for therapeutic effects. However, their pharmacokinetics (PK) and excretion in vivo and the risk of drug-drug interactions (DDI) through inhibiting human cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes remain unclear. METHODS PK and excretion of circulating compounds were investigated in rats using a validated ultra-high-performance liquid chromatography tandem mass spectrometry (UHPLC-MS) method. Further, the inhibitory effects of nine major compounds against eleven CYP and UGT isozymes were assayed using well-accepted specific substrate for each enzyme. RESULTS After dosing, 9 alkaloids were found with C max and t 1/2 values of 0.17-422.70 μmol/L and 1.78-4.33 h, respectively. Additionally, 28 saponins exhibited considerable systemic exposure with t 1/2 values of 0.63-7.22 h, whereas other trace saponins could be negligible or undetected. Besides, over 90% of alkaloids were excreted through hepatobiliary and renal excretion. Likewise, astragalosides and protopanaxatriol (PPT) type ginsenosides also involved in hepatobiliary and/or renal excretion. Protopanaxadiol (PPD) type ginsenosides were mainly excreted to urine. Furthermore, PPD-type ginsenosides were extensively bound (f u-plasma approximately 1%), whereas astragalosides and PPT-type ginsenosides displayed f u-plasma values of 12.35% and 60.23-87.36%, respectively. Moreover, matrine, oxymatrine, astragaloside IV, ginsenoside Rg1, ginsenoside Re, ginsenoside Rd, ginsenoside Rc, and ginsenoside Rb1 exhibited no inhibition or weak inhibition against several common CYP and UGT enzymes IC50 values between 8.81 and 92.21 μM. Through kinetic modeling, their inhibition mechanisms towards those CYP and UGT isozymes were explored with obtained Ki values. In vitro-in vivo extrapolation showed the inhibition of systemic clearance for CYP or UGT substrates seemed impossible due to [I]/Ki no more than 0.1. CONCLUSIONS We summarized the PK behaviors, excretion characteristics and protein binding rates of circulating alkaloids, astragalosides and ginsenosides after intravenous Kang-Ai injection. Furthermore, weak inhibition or no inhibition towards these CYP and UGT activities could not trigger harmful DDI when Kang-Ai injection is co-administered with clinical drugs primarily cleared by these CYP or UGT isozymes.
Collapse
Affiliation(s)
- Zifei Qin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052 China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 China
| | - Mengmeng Jia
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052 China
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052 China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052 China
| | - Zhao Yin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052 China
| | - Zhihong Yao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 China
- College of Pharmacy, Jinan University, Guangzhou, 510632 China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052 China
| | - Xinsheng Yao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 China
- College of Pharmacy, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
9
|
Zhang L, Zhu L, Qu W, Wu F, Hu M, Xie W, Liu Z, Wang C. Insight into tartrate inhibition patterns in vitro and in vivo based on cocrystal structure with UDP-glucuronosyltransferase 2B15. Biochem Pharmacol 2020; 172:113753. [DOI: 10.1016/j.bcp.2019.113753] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/10/2019] [Indexed: 01/08/2023]
|
10
|
Yang J, Li K, He D, Gu J, Xu J, Xie J, Zhang M, Liu Y, Tan Q, Zhang J. Toward a better understanding of metabolic and pharmacokinetic characteristics of low-solubility, low-permeability natural medicines. Drug Metab Rev 2020; 52:19-43. [PMID: 31984816 DOI: 10.1080/03602532.2020.1714646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Today, it is very challenging to develop new active pharmaceutical ingredients. Developing good preparations of well-recognized natural medicines is certainly a practical and economic strategy. Low-solubility, low-permeability natural medicines (LLNMs) possess valuable advantages such as effectiveness, relative low cost and low toxicity, which is shown by the presence of popular products on the market. Understanding the in vivo metabolic and pharmacokinetic characteristics of LLNMs contributes to overcoming their associated problems, such as low absorption and low bioavailability. In this review, the structure-based metabolic reactions of LLNMs and related enzymatic systems, cellular and bodily pharmacological effects and metabolic influences, drug-drug interactions involved in metabolism and microenvironmental changes, and pharmacokinetics and dose-dependent/linear pharmacokinetic models are comprehensively evaluated. This review suggests that better pharmacological activity and pharmacokinetic behaviors may be achieved by modifying the metabolism through using nanotechnology and nanosystem in combination with the suitable administration route and dosage. It is noteworthy that novel nanosystems, such as triggered-release liposomes, nucleic acid polymer nanosystems and PEGylated dendrimers, in addition to prodrug and intestinal penetration enhancer, demonstrate encouraging performance. Insights into the metabolic and pharmacokinetic characteristics of LLNMs may help pharmacists to identify new LLNM formulations with high bioavailability and amazing efficacy and help physicians carry out LLNM-based precision medicine and individualized therapies.
Collapse
Affiliation(s)
- Jie Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Kailing Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Jing Gu
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing, China
| | - Jingyu Xu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jiaxi Xie
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Min Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Yuying Liu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Qunyou Tan
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing, China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Zhang J, Wu C, Gao L, Du G, Qin X. Astragaloside IV derived from Astragalus membranaceus: A research review on the pharmacological effects. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 87:89-112. [PMID: 32089240 DOI: 10.1016/bs.apha.2019.08.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Decoctions prepared from the roots of Astragali Radix are known as "Huangqi" and are widely used in traditional Chinese medicine for treatment of viral and bacterial infections, inflammation, as well as cancer. Astragaloside IV (AS-IV), one of the major compounds from the aqueous extract of Astragalus membranaceus, is a cycloartane-type triterpene glycoside chemical. To date, many studies in cellular and animal models have demonstrated that AS-IV possesses potent protective effects in cardiovascular, lung, kidney and brain. Based on studies over the past several decades, this review systematically summarizes the pharmacological effects, pharmacokinetics and the toxicity of AS-IV. We analyze in detail the pharmacological effects of AS-IV on neuroprotection, liver protection, anti-cancer and anti-diabetes, attributable to its antioxidant, anti-inflammatory, anti-apoptotic properties, and the roles in enhancement of immunity, attenuation of the migration and invasion of cancer cells and improvement of chemosensitivity of chemotherapy drugs. In addition, the latest developments in the combination of AS-IV and other active ingredients of traditional Chinese medicine or chemical drugs are detailed. These pharmacological effects are associated with multiple signaling pathways, including the Raf-MEK-ERK pathway, EGFR-Nrf2 signaling pathway, Akt/PDE3B signaling pathway, AMPK signaling pathway, NF-κB signaling pathway, Nrf2 antioxidant signaling pathways, PI3K/Akt/mTOR signaling pathway, PKC-α-ERK1/2-NF-κB pathway, IL-11/STAT3 signaling pathway, Akt/GSK-3β/β-catenin pathway, JNK/c-Jun/AP-1 signaling pathway, PI3K/Akt/NF-κB pathway, miRNA-34a/LDHA pathway, Nox4/Smad2 pathway, JNK pathway and NF-kB/PPARγ pathway. This review will provide an overall understanding of the pharmacological functions of astragaloside IV on neuroprotection, liver protection, anti-cancer and anti-diabetes. In light of this, AS-IV will be a potent alternative therapeutic agent for treatment of the above mentioned diseases.
Collapse
Affiliation(s)
- Jianqin Zhang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Chuxuan Wu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P. R. China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China.
| |
Collapse
|
12
|
Potential of herb-drug / herb interactions between substrates and inhibitors of UGTs derived from herbal medicines. Pharmacol Res 2019; 150:104510. [DOI: 10.1016/j.phrs.2019.104510] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
|
13
|
Liu YZ, Zhang ZP, Fu ZW, Yang K, Ding N, Hu LG, Fang ZZ, Zhuo X. Per- and polyfluoroalkyl substances display structure-dependent inhibition towards UDP-glucuronosyltransferases. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:113093. [PMID: 31472452 DOI: 10.1016/j.envpol.2019.113093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/31/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a large group of chemicals and can be detected in environmental and human samples all over the world. Toxicity of existing and emerging PFASs will be a long-term source of concern. This study aimed to investigate structure-dependent inhibitory effects of 14 PFASs towards the activity of 11 UDP-glucuronosyltransferase (UGT) isoforms. In vitro UGTs-catalyzed glucuronidation of 4-methylumbelliferone (4-MU) was employed to determine the inhibition of PFASs towards different UGT isoforms. All the PFASs showed <75% of inhibition or stimulation effects on UGT1A3, UGT1A7, UGT1A9, UGT2B4, UGT2B7 and UGT2B17. However, PFASs showed broad inhibition on the activity of UGT1A1 and UGT1A8. The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 μmoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. UGT1A8 was inhibited by 97.6%, 94.8%, 86.3%, 83.4% and 77.1% by PFDA, PFTA, perfluorooctadecanoic acid (PFOcDA), PFDoA and PFOS, respectively. Additionally, PFDA significantly inhibited UGT1A6 and UGT1A10 by 96.8% and 91.6%, respectively. PFDoA inhibited the activity of UGT2B15 by 88.2%. PFDA and PFOS exhibited competitive inhibition towards UGT1A1, and PFDA and PFTA showed competitive inhibition towards UGT1A8. The inhibition kinetic parameter (Ki) were 3.15, 1.73, 13.15 and 20.21 μmoL/L for PFDA-1A1, PFOS-1A1, PFDA-1A8 and PFTA-1A8, respectively. The values were calculated to be 0.3 μmoL/L and 1.3 μmoL/L for the in vivo inhibition of PFDA towards UGT1A1-and UGT1A8-catalyzed metabolism of substances, and 0.2 μmoL/L and 2.0 μmoL/L for the inhibition of PFOS towards UGT1A1 and the inhibition of PFTA towards UGT1A8, respectively. Molecular docking indicated that hydrogen bonds and hydrophobic interactions contributed to the interaction between PFASs and UGT isoforms. In conclusion, exposure to PFASs might inhibit the activity of UGTs to disturb metabolism of endogenous compounds and xenobiotics. The structure-related effects of PFASs on UGTs would be very important for risk assessment of PFASs.
Collapse
Affiliation(s)
- Yong-Zhe Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China
| | - Zhi-Peng Zhang
- Department of Surgery, Peking University Third Hospital, Beijing, China
| | - Zhi-Wei Fu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China
| | - Kun Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China
| | - Ning Ding
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Shaanxi, Xi'an, 710061, China
| | - Li-Gang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China.
| | - Xiaozhen Zhuo
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Shaanxi, Xi'an, 710061, China.
| |
Collapse
|
14
|
Hwang ST, Kim C, Lee JH, Chinnathambi A, Alharbi SA, Shair OHM, Sethi G, Ahn KS. Cycloastragenol can negate constitutive STAT3 activation and promote paclitaxel-induced apoptosis in human gastric cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152907. [PMID: 30981183 DOI: 10.1016/j.phymed.2019.152907] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/25/2019] [Accepted: 03/30/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Cycloastragenol (CAG), a triterpene aglycone is commonly prescribed for treating hypertension, cardiovascular disease, diabetic nephropathy, viral hepatitis, and various inflammatory-linked diseases. HYPOTHESIS We investigated CAG for its action on signal transducer and activator of transcription 3 (STAT3) activation cascades, and its potential to sensitize gastric cancer cells to paclitaxel-induced apoptosis. METHODS The effect of CAG on STAT3 phosphorylation and other hallmarks of cancer was deciphered using diverse assays in both SNU-1 and SNU-16 cells. RESULTS We observed that CAG exhibited cytotoxic activity against SNU-1 and SNU-16 cells to a greater extent as compared to normal GES-1 cells. CAG predominantly caused negative regulation of STAT3 phosphorylation at tyrosine 705 through the abrogation of Src and Janus-activated kinases (JAK1/2) activation. We noted that CAG impaired translocation of STAT3 protein as well as its DNA binding activity. It further decreased cellular proliferation and mediated its anticancer effects predominantly by causing substantial apoptosis rather than autophagy. In addition, CAG potentiated paclitaxel-induced anti-oncogenic effects in gastric tumor cells. CONCLUSIONS Our results indicate that CAG can function to impede STAT3 activation in human gastric tumor cells and therefore it may be a suitable candidate agent for therapy of gastric cancer.
Collapse
Affiliation(s)
- Sun Tae Hwang
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chulwon Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jong Hyun Lee
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Omar H M Shair
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
15
|
Gao WF, Li YX, Zhang WH, Tao R, Yin TT, Wang YJ, Liu LN, Fu ZW, Li SN, Liu NR, Zhang H, Liu G, Zhao LZ, Zhang XP, Zhang CZ. Comparison of the inhibition potential of parthenolide and micheliolide on various UDP-glucuronosyltransferase isoforms. Xenobiotica 2018; 49:1158-1163. [PMID: 30484368 DOI: 10.1080/00498254.2018.1544383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Parthenolide (PTL) and micheliolide (MCL) are sesquiterpene lactones with similar structures, and both of them have been reported to exhibit multiple biochemical and pharmacological activities. This study aims to investigate the inhibition of these two compounds on the activity of UDP-glucuronosyltransferases (UGTs). In vitro incubation mixture for recombinant UGTs-catalyzed glucuronidation metabolism of 4-methylumbelliferone (4-MU) was utilized to investigate the inhibition potential. Inhibition kinetics (including inhibition type and parameters) were determined, and in silico docking was employed to elucidate the inhibition difference between PTL and MCL on UGT1A1. MCL showed no inhibition toward all the UGT isoforms, and PTL showed strong inhibition toward UGT1A1. The half-maximal inhibitory concentration (IC50) of PTL on the activity of UGT1A1 was determined to be 64.4 μM. Inhibition kinetics determination showed that PTL exerted noncompetitive inhibition toward UGT1A1, and the inhibition kinetic constant (Ki) was determined to be 12.1 μM. In silico docking method has been employed to show that hydrogen bonds between PTL and the activity cavity of UGT1A1 contributed to the stronger inhibition of PTL on the activity of UGT1A1 than MCL. In conclusion, PTL can more easily induce drug-drug interaction (DDI) with clinical drugs mainly undergoing UGT1A1-catalyzed glucuronidation.
Collapse
Affiliation(s)
- Wei-Feng Gao
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China
| | - Yi-Xuan Li
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China.,b Department of Clinical Science of Integrated TCM and Western Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Wei-Hua Zhang
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China
| | - Ran Tao
- c Basic Medical College, Hebei North University , Hebei , China
| | - Ting-Ting Yin
- b Department of Clinical Science of Integrated TCM and Western Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Yi-Jia Wang
- d Department of Pathology , Tianjin Union Medical Center , Tianjin , China
| | - Li-Na Liu
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China
| | - Zhi-Wei Fu
- e Department of Toxicology School of Public Health , Tianjin Medical University , Tianjin , China
| | - Sai-Nan Li
- e Department of Toxicology School of Public Health , Tianjin Medical University , Tianjin , China
| | - Nai-Rong Liu
- e Department of Toxicology School of Public Health , Tianjin Medical University , Tianjin , China
| | - Heng Zhang
- f Tianjin Union Medical Center , Tianjin , China
| | - Guang Liu
- f Tianjin Union Medical Center , Tianjin , China
| | - Li-Zhong Zhao
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China
| | - Xi-Peng Zhang
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China
| | - Chun-Ze Zhang
- a Department of Colorectal Surgery , Tianjin Union Medical Center , Tianjin , China
| |
Collapse
|
16
|
Cycloastragenol ameliorates experimental heart damage in rats by promoting myocardial autophagy via inhibition of AKT1-RPS6KB1 signaling. Biomed Pharmacother 2018; 107:1074-1081. [DOI: 10.1016/j.biopha.2018.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 01/17/2023] Open
|
17
|
Yu Y, Zhou L, Yang Y, Liu Y. Cycloastragenol: An exciting novel candidate for age-associated diseases. Exp Ther Med 2018; 16:2175-2182. [PMID: 30186456 PMCID: PMC6122403 DOI: 10.3892/etm.2018.6501] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022] Open
Abstract
Cycloastragenol (CAG) is a triterpenoid saponin compound and a hydrolysis product of the main active ingredient in Astragalus membranaceus (Fisch.) Bunge. An increasing body of evidence has indicated that CAG has a wide spectrum of pharmacological functions, which are attracting attention in the research community. The aim of the present review paper was to review and elucidate the advanced study of CAG. The focus was on advanced studies of CAG in English and Chinese databases; the literature was collected and reviewed to summarize the latest efficacy, pharmacokinetics and adverse reactions of CAG. Extensive pharmacological effects have been attributed to CAG, including telomerase activation, telomere elongation, anti-inflammatory and anti-oxidative properties; CAG has also been reported to improve lipid metabolism. Clinical research has demonstrated that CAG activates telomerase in humans and ameliorates various biomarkers. CAG is absorbed through the intestinal epithelium via passive diffusion and undergoes first-pass hepatic metabolism. Within a certain dose range, oral CAG is relatively safe; however, underlying mechanisms associated with CAG are not clear, and thus, we should be aware of potential adverse reactions associated with CAG. According to existing studies and clinical trials, CAG is safe and has broad application prospects. However, further studies are required to fully understand its efficacy and potential adverse reactions, and to ensure the proper use of CAG is applied to treat diseases clinically.
Collapse
Affiliation(s)
- Yongjie Yu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Limin Zhou
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yuyu Liu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| |
Collapse
|
18
|
You BH, Gong EC, Choi YH. Inhibitory Effect of Sauchinone on UDP-Glucuronosyltransferase (UGT) 2B7 Activity. Molecules 2018; 23:molecules23020366. [PMID: 29425147 PMCID: PMC6017115 DOI: 10.3390/molecules23020366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 01/03/2023] Open
Abstract
Herb-drug interaction (HDI) limits clinical application of herbs and drugs, and inhibition of herbs towards uridine diphosphate (UDP)-glucuronosyltransferases (UGTs) has gained attention as one of the important reasons to cause HDIs. Sauchinone, an active lignan isolated from aerial parts of Saururus chinensis (Saururacease), possesses anti-oxidant, anti-inflammatory, and anti-viral activities. In pharmacokinetics of sauchinone, sauchinone is highly distributed to the liver, forming extensive metabolites of sauchinone via UGTs in the liver. Thus, we investigated whether sauchinone inhibited UGTs to explore potential of sauchinone-drug interactions. In human liver microsomes (HLMs), sauchinone inhibited activities of UGT1A1, 1A3, 1A6, and 2B7 with IC50 values of 8.83, 43.9, 0.758, and 0.279 μM, respectively. Sauchinone also noncompetitively inhibited UGT1A6 and 2B7 with Ki values of 1.08 and 0.524 μM, respectively. In in vivo interaction study using mice, sauchinone inhibited UGT2B7-mediated zidovudine metabolism, resulting in increased systemic exposure of zidovudine when sauchinone and zidovudine were co-administered together. Our results indicated that there is potential HDI between sauchinone and drugs undergoing UGT2B7-mediated metabolism, possibly contributing to the safe use of sauchinone and drug combinations.
Collapse
Affiliation(s)
- Byoung Hoon You
- College of Pharmacy and Intergrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyonggi-do 10326, Korea.
| | - Eun Chae Gong
- College of Pharmacy and Intergrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyonggi-do 10326, Korea.
| | - Young Hee Choi
- College of Pharmacy and Intergrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang, Gyonggi-do 10326, Korea.
| |
Collapse
|
19
|
Comparative analysis of multiple representative components in the herb pair Astragali Radix-Curcumae Rhizoma and its single herbs by UPLC-QQQ-MS. J Pharm Biomed Anal 2018; 148:224-229. [DOI: 10.1016/j.jpba.2017.09.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/08/2017] [Accepted: 09/09/2017] [Indexed: 12/20/2022]
|
20
|
Sun D, Zhang CZ, Ran RX, Cao YF, Du Z, Fu ZW, Huang CT, Zhao ZY, Zhang WH, Fang ZZ. In Vitro Comparative Study of the Inhibitory Effects of Mangiferin and Its Aglycone Norathyriol towards UDP-Glucuronosyl Transferase (UGT) Isoforms. Molecules 2017. [PMID: 28621744 PMCID: PMC6152678 DOI: 10.3390/molecules22061008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mangiferin (MGF), the predominant constituent of extracts of the mango plant Mangifera Indica L., has been investigated extensively because of its remarkable pharmacological effects. In vitro recombinant UGTs-catalyzed glucuronidation of 4-methylumbelliferone (4-MU) was used to investigate the inhibition of mangiferin and aglycone norathyriol towards various isoforms of UGTs in our study, which evaluated the inhibitory capacity of MGF and its aglycone norathyriol (NTR) towards UDP-glucuronosyltransferase (UGT) isoforms. Initial screening experiment showed that deglycosylation of MGF into NTR strongly increased the inhibitory effects towards almost all the tested UGT isoforms at a concentration of 100 μM. Kinetic experiments were performed to further characterize the inhibition of UGT1A3, UGT1A7 and UGT1A9 by NTR. NTR competitively inhibited UGT1A3, UGT1A7 and UGT1A9, with an IC50 value of 8.2, 4.4, and 12.3 μM, and a Ki value of 1.6, 2.0, and 2.8 μM, respectively. In silico docking showed that only NTR could dock into the activity cavity of UGT1A3, UGT1A7 and UGT1A9. The binding free energy of NTR to UGT1A3, 1A7, 1A9 were −7.4, −7.9 and −4.0 kcal/mol, respectively. Based on the inhibition evaluation standard ([I]/Ki < 0.1, low possibility; 0.1 < [I]/Ki < 1, medium possibility; [I]/Ki > 1, high possibility), an in vivo herb–drug interaction between MGF/NTR and drugs mainly undergoing UGT1A3-, UGT1A7- or UGT1A9-catalyzed metabolism might occur when the plasma concentration of NTR is above 1.6, 2.0 and 2.8 μM, respectively.
Collapse
Affiliation(s)
- Dan Sun
- College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Chun-Ze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China.
| | - Rui-Xue Ran
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Yun-Feng Cao
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
| | - Zuo Du
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Zhi-Wei Fu
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Chun-Ting Huang
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Zhen-Ying Zhao
- Tianjin Union Medical Center, 190 Jieyuan Road, Hongqiao District, Tianjin 300121, China.
| | - Wei-Hua Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China.
| | - Zhong-Ze Fang
- Key Laborotary of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou 121001, Liaoning, China.
- Department of Toxicology, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| |
Collapse
|