1
|
Tan Y, Zhang F, Fan X, Lu S, Liu Y, Wu Z, Huang Z, Wu C, Cheng G, Li B, Huang J, Stalin A, Zhou W, Wu J. Exploring the effect of Yinzhihuang granules on alcoholic liver disease based on pharmacodynamics, network pharmacology and molecular docking. Chin Med 2023; 18:52. [PMID: 37165407 PMCID: PMC10173499 DOI: 10.1186/s13020-023-00759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Yinzhihuang granules (YZHG) is a commonly used Chinese patent medicine for the treatment of liver disease. However, the mechanism of YZHG in alcoholic liver disease (ALD) is still unclear. METHODS This study combined liquid chromatography-mass spectrometry technology, pharmacodynamics, network pharmacology and molecular docking methods to evaluate the potential mechanism of YZHG in the treatment of ALD. RESULTS A total of 25 compounds including 4-hydroxyacetophenone, scoparone, geniposide, quercetin, baicalin, baicalein, chlorogenic acid and caffeic acid in YZHG were identified by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). The pharmacodynamic investigations indicated that YZHG could improve liver function and the degree of liver tissue lesions, and reduce liver inflammation and oxidative stress in ALD mice. Network pharmacology analysis showed that YZHG treated ALD mainly by regulating inflammation-related signaling pathways such as the PI3K-Akt signaling pathway. The results of the PPI network and molecular docking showed that the targets of SRC, HSP90AA1, STAT3, EGFR and AKT1 could be the key targets of YZHG in the treatment of ALD. CONCLUSION This study explored the potential compounds, potential targets and signaling pathways of YZHG in the treatment of ALD, which is helpful to clarify the efficacy and mechanism of YZHG and provide new insights for the clinical application of YZHG.
Collapse
Affiliation(s)
- Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fanqin Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guoliang Cheng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Bing Li
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jiaqi Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| | - Wei Zhou
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Wang XB, Wang ML, Chu YJ, Zhou PP, Zhang XY, Zou J, Zuo LH, Shi YY, Kang J, Li B, Cheng WB, Sun Z, Zhang XJ, Du SZ. Integrated pharmacokinetics and pharmacometabolomics to reveal the synergistic mechanism of a multicomponent Chinese patent medicine, Mailuo Shutong pills against thromboangiitis obliterans. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154709. [PMID: 36774843 DOI: 10.1016/j.phymed.2023.154709] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/23/2023] [Accepted: 02/05/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Mailuo Shutong Pills (MLST) have displayed pharmacological activity against thromboangiitis obliterans (TAO). However, the active ingredients and therapeutic mechanism of MLST against TAO remained to be further clarified. PURPOSE The aim of this study was to explore the active components of MLST and their synergistic mechanism against TAO by integrating pharmacokinetics (PK) and pharmacometabolomics (PM). METHODS TAO model rats were established by sodium laurate solution. Firstly, the efficacy of MLST was evaluated by gangrene score, blood flow velocity, and hematoxylin-eosin (H&E) staining. Secondly, PK research was conducted on bioavailable components to characterize their dynamic behaviors under TAO. Thirdly, multiple plasma and urine metabolic biomarkers for sodium laurate-induced TAO rats were found by untargeted metabolomics, and then variations in TAO-altered metabolites following MLST treatment were analyzed utilizing multivariate and bioinformatic analysis. Additionally, metabolic pathway analysis was performed using MetaboAnalyst. Finally, the dynamic link between absorbed MLST-compounds and TAO-associated endogenous metabolites was established by correlation analysis. RESULTS MLST significantly alleviated gangrene symptoms by improving the infiltration of inflammatory cells and blood supply in TAO rats. Significant differences in metabolic profiles were found in 17 differential metabolites in plasma and 24 in urine between Sham and TAO rats. The 10 bioavailable MLST-compounds, such as chlorogenic acid and paeoniflorin, showed positive or negative correlations with various TAO-altered metabolites related to glutamate metabolism, histidine metabolism, arachidonic acid metabolism and so on. CONCLUSION This study originally investigated the dynamic interaction between MLST and the biosystem, providing unique insight for disclosing the active components of MLST and their synergistic mechanisms against TAO, which also shed light on new therapeutic targets for TAO and treatment.
Collapse
Affiliation(s)
- Xiao-Bao Wang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Meng-Li Wang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Yao-Juan Chu
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Pei-Pei Zhou
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Xiang-Yu Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Jing Zou
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Li-Hua Zuo
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Ying-Ying Shi
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Jian Kang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China
| | - Bing Li
- State Key Laboratory of Common Technology of Traditional Chinese Medicine and Pharmaceuticals, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Wen-Bo Cheng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Zhi Sun
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China.
| | - Xiao-Jian Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China.
| | - Shu-Zhang Du
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Jianshe East Road 1, Zhengzhou 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, Jianshe East Road 1, Zhengzhou 450052, China.
| |
Collapse
|
3
|
Yisimayili Z, Chao Z. A review on phytochemicals, metabolic profiles and pharmacokinetics studies of the different parts (juice, seeds, peel, flowers, leaves and bark) of pomegranate (Punica granatum L.). Food Chem 2022; 395:133600. [DOI: 10.1016/j.foodchem.2022.133600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 11/04/2022]
|
4
|
Liu F, Zeng Y, Dai P, Huang K, Zhang K, Tao T, Wang M, Zhu C, Lin C. Comparative Pharmacokinetics of Three Bioactive Diterpenoids of Rabdosia serra Extract in Normal and Con A-Induced Liver Injury Rats Using UPLC-MS/MS. Front Pharmacol 2022; 13:944949. [PMID: 35903341 PMCID: PMC9323086 DOI: 10.3389/fphar.2022.944949] [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: 05/16/2022] [Accepted: 06/07/2022] [Indexed: 12/18/2022] Open
Abstract
Rabdosia serra (Maxim.) Hara (R. serra), one of the source plants of “Xihuangcao”, has been widely used as a Chinese folk herb with the concomitant function of both medicine and foodstuff for the prevention and treatment of liver disease. Diterpenoids were considered as the major bioactive components in R. serra, responsible for their effect on hepatoprotection in previous phytochemical and pharmacological studies, while few comparative pharmacokinetic studies have been conducted under the physiological and pathological conditions. To reveal the difference in the pharmacokinetics process of R. serra extract (RSE) in normal and Con A-induced liver injury rats, a rapid ultra-high-pressure liquid chromatography–tandem mass spectrometry method (total running time: 5 min) was established to simultaneously determine three bioactive diterpenoids (enmein, epinodosin, and isodocarpin) in rat plasma. The results showed significant differences in the pharmacokinetic properties of three analytes between the physiological and pathological states. Compared with normal rats, the AUC of the three analytes was remarkably higher in liver injury rats, while the Tmax, T1/2, and MRT were shortened. It indicated that RSE has higher exposure and quicker elimination in liver injury rats than that in normal rats. Our results suggested that the pharmacokinetics of hepatoprotective medications was affected by liver injury, which prospected to provide essential information for guiding the healthcare and clinical application of R. serra in pathological states.
Collapse
Affiliation(s)
- Fangle Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Zeng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengyu Dai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaiwen Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaihui Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tao Tao
- Guangzhou Chest Hospital, Guangzhou, China
| | - Meiqi Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chenchen Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chaozhan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Wang Z, Liu H, Cao Y, Zhang T, Guo H, Wang B. A novel method for investigating the mechanism of anti-rheumatoid arthritis activity of Angelicae pubescentis Radix by integrating UHPLC-QTOF/MS and network pharmacology. Biomed Chromatogr 2022; 36:e5389. [PMID: 35484722 DOI: 10.1002/bmc.5389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 11/09/2022]
Abstract
The study aimed to establish a strategy to elucidate the in vivo constituents of Angelicae pubescentis Radix (APR, also known as Duhuo) and reveal the probable mechanisms underlying the anti-rheumatoid arthritis activity of APR. APR, first recorded in Shennong Bencao Jing, is mainly used to treat Bi syndrome. Eleven absorbed components of APR were successfully identified using the Rheumatoid arthritis (RA) rat model and the UHPLC-QTOF/MS technique. Two active ingredients (osthole, and columbianadin) and five corresponding targets (PTGS1, PTGS2, RXRA, CCNA2 and ACHE) were found to construct a compound-protein interaction network in RA. In addition, a non-alcoholic fatty liver disease (NAFLD) pathway, which was related to anti-RA activity, was eventually identified by KEGG analysis. Subsequently, molecular docking was performed by establishing a mixed matrix network including the absorbed component, corresponding target, and signaling pathway with two key compounds (osthole and columbianadin) and two important targets (PTGS2 and PTGS1). The result of molecular docking is in agreement with the network pharmacology (NP).
Collapse
Affiliation(s)
- Zhen Wang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Hui Liu
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Yunxiang Cao
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, China
| | - Tiantian Zhang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Hongyan Guo
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Bin Wang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| |
Collapse
|
6
|
Nabi F, Tao W, Ye R, Li Z, Lu Q, Shang Y, Hu Y, Fang J, Bhutto ZA, Liu J. Penthorum Chinense Pursh Extract Alleviates Aflatoxin B1-Induced Liver Injury and Oxidative Stress Through Mitochondrial Pathways in Broilers. Front Vet Sci 2022; 9:822259. [PMID: 35187148 PMCID: PMC8847786 DOI: 10.3389/fvets.2022.822259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Aflatoxin is an important toxicant of the fungal origin and poses a threat to the poultry industry. This study was designed to reveal the underlying mechanism and protective methods against aflatoxin B1 (AFB1)-induced liver injury, oxidative stress, and apoptosis using a Traditional Chinese medicine, Penthorum chinense Pursh extract (PCPE), in broilers. A total of 164 (day-old) broilers were equally allocated to the control, AFB1 (3 mg/kg feed), positive drug (Yin-Chen-Hao Tang extract, 10 ml/kg feed), PCPE (2 g PCPE/kg), and PCPE low, medium, and high dose groups (1 g, 2 g, 3 g PCPE/kg feed, respectively). AFB1 significantly decreased the growth performance and serum immunoglobulin level, altered normal serum biochemical parameters and antioxidant activities, and induced histopathological lesions in the liver as compared to control group. Additionally, AFB1 significantly up-regulated the mRNA expression levels of apoptosis-related genes such as Bax, Bak, caspase-9, caspase-3, and p53, whereas it down-regulated the expression levels of BCL2 in the liver of broilers. The supplementation of different doses of PCPE to AFB1-affected birds significantly eased AFB1 negative effects by improving growth performance, immunoglobulin level, and oxidative capacity, and reversed oxidative stress and pathological lesions in liver. Furthermore, supplementation of PCPE to the AFB1 group reversed apoptosis by significantly down-regulating the mRNA expression levels of Bax, Bak, caspase-9, caspase-3, and p53 and up-regulating the expression levels of BCL2 in the liver of broilers. Based on these results, we conclude that supplementation of PCPE is protective and safe against oxidative stress, is anti-apoptotic, and reverses the liver damage caused by AFB1 in broilers.
Collapse
Affiliation(s)
- Fazul Nabi
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
- Department of Poultry Science, Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Pakistan
| | - Weilai Tao
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Ruiling Ye
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenzhen Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qin Lu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yangfei Shang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Hu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jiali Fang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zohaib Ahmed Bhutto
- Department of Poultry Science, Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Pakistan
| | - Juan Liu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
- Chinese Veterinary Herbal Drugs Innovation Research Laboratory, University Veterinary Science Engineering Research Center in Chongqing, Chongqing, China
- Immunology Research Center of Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Juan Liu
| |
Collapse
|
7
|
Tao W, Li Z, Nabi F, Hu Y, Hu Z, Liu J. Penthorum chinense Pursh Compound Ameliorates AFB1-Induced Oxidative Stress and Apoptosis via Modulation of Mitochondrial Pathways in Broiler Chicken Kidneys. Front Vet Sci 2021; 8:750937. [PMID: 34692815 PMCID: PMC8531719 DOI: 10.3389/fvets.2021.750937] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a carcinogenic mycotoxin widely present in foods and animal feeds; it represents a great risk to human and animal health. The aim of this study was to investigate the protective effects of Penthorum chinense Pursh compound (PCPC) against AFB1-induced damage, oxidative stress, and apoptosis via mitochondrial pathways in kidney tissues of broilers. One-day-old chickens (n = 180) were randomly allocated to six groups: control, AFB1 (2.8 mg AFB1/kg feed), positive drug (10 mLYCHT/kg feed), and PCPC high, medium, and low-dose groups (15, 10, and 5 ml PCPC/kg feed, respectively). AFB1 treatment reduced weight gain and induced oxidative stress and kidney damage in broiler tissues; however, PCPC supplementation effectively enhanced broiler performance, ameliorated AFB1-induced oxidative stress, and inhibited apoptosis in the kidneys of broilers. The mRNA expression levels of mitochondria-related apoptosis genes (Bax, Bak, cytochrome c, caspase-9, and caspase-3) were significantly increased, whereas BCL2 expression level decreased in the AFB1 group. Supplementation of PCPC to the AFB1 group significantly reversed the changes in mRNA expression levels of these apoptosis-associated genes compared to those in the AFB1 group. The mRNA levels of NRF2 and HMOX1 in the kidneys of the AFB1 group were significantly reduced compared to those in the control group, whereas PCPC significantly increased the NRF2 and HMOX1 mRNA levels. AFB1 decreased the levels of Beclin1, LC3-I, and LC3-II and increased P53 levels in the kidney compared to those in the control, whereas PCPC significantly reversed these changes to normal levels of autophagy-related genes compared to those in the AFB1 group. In conclusion, our findings demonstrated that PCPC ameliorated AFB1-induced oxidative stress by regulating the expression of apoptosis-related genes and mitochondrial pathways. Our results suggest that PCPC represents a natural and safe agent for preventing AFB1-induced injury and damage in broiler tissues.
Collapse
Affiliation(s)
- Weilai Tao
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenzhen Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Fazul Nabi
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Hu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zeyu Hu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Juan Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Chinese Veterinary Herbal Drugs Innovation Research Lab, University Veterinary Science Engineering Research Center in Chongqing, Chongqing, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
8
|
Wang Y, Li Y, Zhang H, Zhu L, Zhong J, Zeng J, Meng C, Wu J, Wang T, Shi R, Yuan W, Jiang J, Liu P, Ma Y. Pharmacokinetics-based comprehensive strategy to identify multiple effective components in Huangqi decoction against liver fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153513. [PMID: 33647776 DOI: 10.1016/j.phymed.2021.153513] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/20/2021] [Accepted: 02/11/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Huangqi decoction (HQD) has been used to treat chronic liver diseases since the 11th century, but the effective components in HQD against liver fibrosis have not been definitively clarified. PURPOSE To investigate and identify multiple effective components in HQD against liver fibrosis using a pharmacokinetics-based comprehensive strategy. METHODS The absorbed representative components in HQD and their metabolites were detected in human plasma and urine using high-resolution mass spectrometry combined with a database-directed method, and then pharmacokinetics in multiple HQD components in human plasma was analyzed by ultra-performance liquid chromatography coupled with triple-quadruple mass spectrometry. Furthermore, the anti-fibrotic effect of potential effective HQD components was studied in LX-2 cells and that of a multi-component combination of HQD (MCHD) was verified in a mouse CCl4-induced hepatic fibrosis model. RESULTS Twenty-four prototype components in HQD and 17 metabolites were identified in humans, and the pharmacokinetic characteristics of 14 components were elucidated. Among these components, astragaloside IV, cycloastragenol, glycyrrhizic acid, glycyrrhetinic acid, liquiritigenin, and isoliquiritigenin downregulated the mRNA expression of α-SMA; cycloastragenol, calycosin-7-O-β-D-glucoside, formononetin, glycyrrhetinic acid, liquiritin, and isoliquiritin downregulated the mRNA expression of Col I; and calycosin, liquiritigenin, isoliquiritigenin, cycloastragenol, and glycyrrhetinic accelerated the apoptosis of LX-2 cells. MCHD reduced serum aminotransferase activity and hepatic collagen fibril deposition in mice with CCl4-induced hepatic fibrosis. CONCLUSION Using the pharmacokinetics-based comprehensive strategy, we revealed that multiple effective HQD components act together against liver fibrosis.
Collapse
Affiliation(s)
- Yahang Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201204, China; E-Institute of Shanghai Municipal Education Committee, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leilei Zhu
- GCP center, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Zhong
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiakai Zeng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Cong Meng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rong Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weian Yuan
- GCP center, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian Jiang
- GCP center, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201204, China; E-Institute of Shanghai Municipal Education Committee, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
9
|
Bridi R, Lino von Poser G, Gómez M, Andia ME, Oyarzún JE, Núñez P, Vasquez Arias AJ, Espinosa-Bustos C. Hepatoprotective species from the Chilean medicinal flora: Junellia spathulata (Verbenaceae). JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113543. [PMID: 33152429 DOI: 10.1016/j.jep.2020.113543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chilean population relies on medicinal plants for treating a wide range of illnesses, especially those of the gastrointestinal system. Junellia spathulata (Gillies & Hook.) Moldenke var. spathulata (Verbenaceae), called as "verbena-azul-de-cordilleira", is a medicinal plant native to Argentina and Chile traditionally used for treating digestive disorders. Although the species of the genus are important as therapeutic resources for the Andean population, the plants are very scarcely studied. AIMS OF THE STUDY The purpose of the present study was to find out the main constituents and investigate the protective effect of J. spathulata against oxidative stress induced by the potent oxidant 2,2'-azobis (2-amidinopropane) dihydrochloride (AAPH) in human hepatoblastoma cells. MATERIALS AND METHODS The crude methanol extract of J. spathulata and an iridoid obtained by chromatographic processes were tested to access the hepatoprotective effect and cytotoxicity in HepG2 cell. In addition, the reducing power of the samples and their ability to scavenge free radicals were evaluated using FRAP and ORAC assay systems. RESULTS The iridoid asperuloside, the main compound of the crude methanol extract of J. spathulata, was isolated and identified by means of NMR analysis. The crude methanol extract of J. spathulata and asperuloside protected HepG2 cells against oxidative damage triggered by AAPH-derived free radicals. This effect can be credited to the ability of the extract and asperuloside to protect the liver cells from chemical-induced injury, which might be correlated to their free radical scavenging potential. CONCLUSIONS This study experimentally evidenced the ethnopharmacological usefulness of J. spathulata as a treatment of digestive disorders. Our result could stimulate further investigations of hepatoprotective agents in other Chilean Junellia species.
Collapse
Affiliation(s)
- Raquel Bridi
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna 4860, Macul, Santiago, 702843, Chile.
| | - Gilsane Lino von Poser
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia - UFRGS, Porto Alegre, Brazil
| | - Miguel Gómez
- Departamento de Ciencias Vegetales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Chile
| | - Marcelo E Andia
- Departamento de Radiología y Centro de Imágenes Biomédicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile
| | - Juan Esteban Oyarzún
- Departamento de Radiología y Centro de Imágenes Biomédicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile
| | - Paula Núñez
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna 4860, Macul, Santiago, 702843, Chile
| | - Ariadsna Jael Vasquez Arias
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna 4860, Macul, Santiago, 702843, Chile
| | - Christian Espinosa-Bustos
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna 4860, Macul, Santiago, 702843, Chile
| |
Collapse
|
10
|
Liu Y, Mapa MST, Sprando RL. Anthraquinones inhibit cytochromes P450 enzyme activity in silico and in vitro. J Appl Toxicol 2021; 41:1438-1445. [PMID: 33438235 DOI: 10.1002/jat.4134] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/29/2020] [Accepted: 12/14/2020] [Indexed: 01/11/2023]
Abstract
Anthraquinones exhibit various pharmacological activities (e.g., antioxidant and laxative) and are commonly found in consumer products including foods, dietary supplements, drugs, and traditional medicines. Despite their widespread use, there are limited data available on their toxicokinetic properties. Cytochrome P450 enzymes (CYPs) in the liver play major roles in metabolizing exogenous chemicals (e.g., pharmaceuticals, food ingredients, and environmental pollutants) and endogenous biomolecules (e.g., steroid hormones and cholesterol). Inhibition of CYP activities may lead to serious interactions among these compounds. Here, in silico (quantitative structure-activity relationship modeling) and in vitro (human recombinant enzymes and liver microsomes) methods were used to identify inhibitors of five major CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) among 22 anthraquinones. First, in silico prediction and in vitro human recombinant enzyme assays were conducted for all compounds, and results showed that most of the anthraquinones were potent CYP1A2 inhibitors. Second, five selected anthraquinones (emodin, aloe-emodin, rhein, purpurin, and rubiadin) were further evaluated in human liver microsomes. Finally, plasma concentrations of the five anthraquinones in animal and humans were identified in the literature and compared to their in vitro inhibition potency (IC50 values) towards CYP activities. Emodin, rhein, and aloe-emodin inhibited activities of multiple CYPs in human liver microsomes and potential in vivo inhibition may occur due to their high plasma concentrations. These in silico and in vitro results enabled rapid identification of potential CYP inhibitors and prioritized future in-depth studies.
Collapse
Affiliation(s)
- Yitong Liu
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Mapa S T Mapa
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, USA
| |
Collapse
|
11
|
Tsai FJ, Yang PY, Chen CJ, Li JP, Li TM, Chiou JS, Cheng CF, Chuang PH, Lin TH, Liao CC, Huang SM, Ban B, Liang WM, Lin YJ. Decreased overall mortality rate with Chinese herbal medicine usage in patients with decompensated liver cirrhosis in Taiwan. BMC Complement Med Ther 2020; 20:221. [PMID: 32664975 PMCID: PMC7362535 DOI: 10.1186/s12906-020-03010-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background Liver cirrhosis is one of the main causes of the morbidity and mortality in liver diseases. Chinese herbal medicine (CHM) has long been used for the clinical treatment of liver diseases. This study was designed to explore the usage frequency and prescription patterns of CHM for patients with decompensated liver cirrhosis and to evaluate the long-term effects of CHM on overall mortality. Methods Two thousand four hundred sixty-seven patients with decompensated liver cirrhosis (ICD-9-CM code: 571.2, 571.5, and 571.6) diagnosed between 2000 and 2009 in Taiwan were identified from the registry for catastrophic illness patients. Of these, 149 CHM users and 298 CHM non-users were matched for age, gender, and Charlson comorbidity index score. The chi-squared test, paired Student’s t-test, Cox proportional hazard model, and Kaplan–Meier method were applied for various comparisons between these groups of patients. Results CHM-treated patients showed a lower overall mortality risk compared with non-treated patients (Multivariable: p < 0.0001; HR: 0.54, 95% CI: 0.42–0.69). The cumulative incidence of overall mortality was lower in the CHM-treated group (stratified log-rank test, p = 0.0002). The strongest CHM co-prescription pattern- Yin-Chen-Hao-Tang (YCHT) → Long-Dan-Xie-Gan-Tang (LDXGT) had the highest support, followed by Zhi-Zi (ZZ) → Yin-Chen-Wu-Ling-San (YCWLS) and Bai-Hua-She-She-Cao (BHSSC) → Da-Huang (DaH). Conclusion CHM, as adjunct therapy, might decrease the risk of overall mortality in patients with decompensated liver cirrhosis. CHM co-prescription patterns and network analysis showed that comprehensive herbal medicines have a protective role against liver fibrosis. Further studies are required to enhance the knowledge of safety and efficacy of CHM in patients with decompensated liver cirrhosis.
Collapse
Affiliation(s)
- Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan.,Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Asia University, Taichung, Taiwan
| | - Pei-Yuu Yang
- Department of Traditional Chinese Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chao-Jung Chen
- Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Ju-Pi Li
- School of Chinese Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan.,Rheumatism Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Te-Mao Li
- School of Chinese Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan
| | - Jian-Shiun Chiou
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan
| | - Chi-Fung Cheng
- Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biostatistics, School of Public Health, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan
| | - Po-Heng Chuang
- Division of Hepato-gastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ting-Hsu Lin
- Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Chu Liao
- Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Shao-Mei Huang
- Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Bo Ban
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, China
| | - Wen-Miin Liang
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan.
| | - Ying-Ju Lin
- School of Chinese Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan. .,Genetic Center, Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
12
|
Hu Y, Zeng H, Huang J, Jiang L, Chen J, Zeng Q. Traditional Asian Herbs in Skin Whitening: The Current Development and Limitations. Front Pharmacol 2020; 11:982. [PMID: 32733239 PMCID: PMC7358643 DOI: 10.3389/fphar.2020.00982] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
In Asia, the market for whitening cosmetics is expanding rapidly, more and more people prefer to use natural products. Driven by natural product demand and technical advances, herbal research is also developing fast. Lots of studies reported that Asian herbal reagents can reduce melanogenesis, these findings provide evidence for the whitening application of Asian herbs. However, the current development status and challenges of herbal research need attention too. By reviewing these studies, different problems in studying herbal formulas, extracts, and active ingredients were presented. One of the most influential troubles is that the components of herbs are too complex to obtain reliable results. Thus, an understanding of the overall quality of herbal research is necessary. Further, 90 most cited Asian herbal studies on whitening were collected, which were conducted between 2017 and 2020, then statistical analysis was carried out. This work provided a comprehensive understanding of Asian herbal research in skin whitening, including the overall status and quality, as well as the focuses and limitations of these studies. By proactively confronting and analyzing these issues, it is suggested that the focus of herbal medicine research needs to shift from quantity to quality, and the new stage of development should emphasize transformation from research findings to whitening products.
Collapse
Affiliation(s)
- Yibo Hu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongliang Zeng
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China
| | - Jinhua Huang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Liu H, Sun Z, Tian X, Feng Q, Guo Z, Chen S, Yin H, Wang Y, Xu Z, Xie L, Hu P, Huang C. Systematic investigation on the chemical basis of anti-NAFLD Qushi Huayu Fang. Part 1: A study of metabolic profiles in vivo and in vitro by high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry. Biomed Chromatogr 2020; 34:e4805. [PMID: 32012315 DOI: 10.1002/bmc.4805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/15/2020] [Accepted: 01/30/2020] [Indexed: 12/11/2022]
Abstract
Qushi Huayu Fang (QHF) is a clinic-empirical prescription for treating non-alcoholic fatty liver disease (NAFLD) in China, which is composed of five herbs. However, the bioactive constituents responsible for the efficacy of QHF remain unclear. Thus, a high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry method was established and adopted to identify the constituents of QHF, and profile its metabolism in vivo and in vitro. Among the 66 constituents in QHF, only 14 compounds of six structural types were absorbed, and 34 metabolites were generated through eight metabolic pathways. A total of 20 metabolites were first reported, including four organic acids, one iridoid, two flavones, five naphthols, three anthraquinones, and five stilbenes. Glucuronidation and sulfation were the main metabolic pathways, and the intestinal metabolism played an important role in the biotransformation of QHF. Many compounds, especially those detected in the liver, the target organ of QHF, were reported to display the anti-NAFLD activity. This is the first study to explore the constituents of QHF and its metabolism in vivo and in vitro, thus realizing the first step to clarify the chemical basis of QHF qualitatively, and laying the foundation for further research on the anti-NAFLD mechanism of QHF.
Collapse
Affiliation(s)
- Huan Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhaolin Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoting Tian
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qin Feng
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziqiong Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuoji Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hao Yin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yangyang Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhou Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Like Xie
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Pei Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chenggang Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Genipin Ameliorates Carbon Tetrachloride-Induced Liver Injury in Mice via the Concomitant Inhibition of Inflammation and Induction of Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3729051. [PMID: 31885784 PMCID: PMC6927019 DOI: 10.1155/2019/3729051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 01/07/2023]
Abstract
Genipin, as the most effective ingredient of various traditional medications, encompasses antioxidative, anti-inflammatory, and antibacterial capacities. More recently, it is suggested that genipin protects against septic liver damage by restoring autophagy. The purpose of the current study was to explore the protective effect of genipin against carbon tetrachloride- (CCl4-) induced acute liver injury (ALI) and its underlying molecular machinery. Our results indicated that treatment with genipin significantly reduced CCl4-induced hepatotoxicity by ameliorating histological liver changes, decreasing the aspartate aminotransferase and alanine transaminase levels, alleviating the secretion of inflammatory cytokines, and promoting autophagic flux. Moreover, genipin effectively induced the conversion of LC3 and inhibition of p62 accumulation. The liver expressions of ATG5, ATG7, and ATG12 were significantly increased by genipin pretreatment in the ALI mice model. This protective effect may be mediated by the inhibition of mTOR and the activation of p38 MAPK signaling pathways. Meanwhile, genipin attenuated CCl4-induced inflammatory response by inhibiting the NF-κB and STAT3 signaling pathway. In addition, pretreatment with autophagy inhibitor 3-methyladenine (3-MA) or inhibition of p38 MAPK by SB203580 abolished the hepatoprotective effect of genipin. Taken together, our study implicates that genipin has a protective potential against CCl4-induced hepatotoxicity, which might be strongly associated with the induction of autophagy and the attenuation of inflammatory response.
Collapse
|
15
|
Zhong M, Tian X, Chen S, Chen M, Guo Z, Zhang M, Zheng G, Li Z, Shi Z, Wang G, Gao H, Liu F, Huang C. Identifying the active components of Baihe-Zhimu decoction that ameliorate depressive disease by an effective integrated strategy: a systemic pharmacokinetics study combined with classical depression model tests. Chin Med 2019; 14:37. [PMID: 31572489 PMCID: PMC6757420 DOI: 10.1186/s13020-019-0254-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background Modern pharmacological studies have demonstrated that Baihe–Zhimu decoction (BZD) has antidepressant effects. However, the complex composition and lack of clear evaluation standards for BZD make it less likely to be understood and accepted than evidence-based active natural compounds. Methods In this study, an effective method for the identification of antidepressant components was demonstrated and applied to BZD. The first step was to evaluate the efficacy of BZD by the forced swimming test (FST) and the tail suspension test (TST), followed by successive quantitative analyses of the absorbed constituents at different stages, such as before hepatic disposition, liver distribution, after hepatic disposition and brain distribution after the oral administration of BZD. Finally, the compounds detected in the brain were confirmed by activity testing. Results Our investigation observed that timosaponin BII and timosaponin BIII were accurately determined in the brain after oral administration of BZD, and they were further confirmed to reduce the immobility time in the FST and TST. As described above, timosaponin BII and timosaponin BIII were used to scientifically and reasonably explain the effective chemical basis of the effect of BZD on depression. Conclusions This research affords an effective method to discover lead molecules for antidepressants from traditional Chinese medicine.
Collapse
Affiliation(s)
- Ming Zhong
- 1College of Pharmacy, Jining Medical University, Rizhao, 276826 People's Republic of China
| | - Xiaoting Tian
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Shuoji Chen
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Mingcang Chen
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Ziqiong Guo
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Minna Zhang
- 1College of Pharmacy, Jining Medical University, Rizhao, 276826 People's Republic of China
| | - Gongpu Zheng
- 1College of Pharmacy, Jining Medical University, Rizhao, 276826 People's Republic of China
| | - Zhixiong Li
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Zhangpeng Shi
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Guanghui Wang
- 1College of Pharmacy, Jining Medical University, Rizhao, 276826 People's Republic of China
| | - Honggang Gao
- 1College of Pharmacy, Jining Medical University, Rizhao, 276826 People's Republic of China
| | - Fang Liu
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| | - Chenggang Huang
- 2Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203 People's Republic of China
| |
Collapse
|
16
|
Yan Y, Jun C, Lu Y, Jiangmei S. Combination of metformin and luteolin synergistically protects carbon tetrachloride-induced hepatotoxicity: Mechanism involves antioxidant, anti-inflammatory, antiapoptotic, and Nrf2/HO-1 signaling pathway. Biofactors 2019; 45:598-606. [PMID: 31336028 DOI: 10.1002/biof.1521] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022]
Abstract
Liver diseases are one of the fatal disorders due to the vital role of the liver. Carbon tetrachloride (CCl4 ) is the most perceived chemical substance utilized in developing models of hepatic damage. Metformin (Met) is a potent antidiabetic and redox modulatory agent that has shown anticancer and protective effects on various organs. Therefore, addition of therapy with natural antioxidative agents or herbal extracts shows defensive impacts against different injuries inside the body. Luteolin (Lut) can be found in several customary Chinese remedies. It has been reported for various pharmacological actions such as antitumor, antioxidative, and anti-inflammatory impacts. Here, the liver injury rat model was established using CCl4 (1.00 mL/kg body weight) in vivo. The protective roles of Met and Lut separately or in combination were observed in hepatotoxicity induced by CCl4 . The result was shown that both Met and Lut, while individually used, were normally active in diminishing CCl4 -caused hepatotoxicity. The combination of two drugs performed synergistically to improve liver damage caused by CCl4 , as shown by the considerably improved liver dysfunction. Met and Lut showed highly antioxidative effects on CCl4 -treated rats moderately by increasing the activities and expression of the antioxidant enzymes. Along with this, a combination of Met and Lut significantly suppressed inflammatory responses, which is evidenced by the reduced level of inflammatory cytokines together with interleukin 1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and interleukin 6 (IL-6). Additionally, CCl4 -agitated apoptosis was intensely reduced by Met and Lut through reducing cleaved caspase-3 and Bax (pro-apoptotic factor) while increasing Bcl-2 (antiapoptotic factor) signaling pathways. Cotreatments of Met and Lut upregulated nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) expression in the CCl4 -intoxicated rat's liver. The above result recommended that combination of Met and Lut may have a substantial potential and synergizing impact against CCl4 -induced hepatotoxicity.
Collapse
Affiliation(s)
- Yang Yan
- Department of Digestive Medicine, Hefei Second People's Hospital, Hefei, China
| | - Chen Jun
- Department of Digestive Medicine, Hefei Second People's Hospital, Hefei, China
| | - Yang Lu
- Department of Digestive Medicine, Hefei Second People's Hospital, Hefei, China
| | - Song Jiangmei
- Department of Internal Medicine, CAS Cancer Hospital, Hefei, China
| |
Collapse
|