1
|
Zhou SK, Xu JD, Gao XQ, Zhang RJ, Cheng FF, Yao WF, Zhang Y, Geng T, Zhang L. Fructus Jujubae cooperated with water-expelling members in Shizao decoction alleviated intestinal injury and malignant ascites by modulating gut microbiota and metabolic homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155895. [PMID: 39084184 DOI: 10.1016/j.phymed.2024.155895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/11/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Shizao decoction (SZD) consisted of Euphorbia kansui (EK), Euphorbia pekinensis (EP), Daphne genkwa (DG), and Fructus Jujubae (FJ) is a classic Chinese herbal medicine formula for treating malignant ascites, which is closely related to the modulation of gut microbiota by our previous study. For water-expelling members (WEM) including EK, EP, and DG may have side effects on the intestine, FJ is employed for detoxification and effectivity enhancement of WEM. However, the underlying mechanism for the compatibility of WEM and FJ is still unknown. PURPOSE To investigate the effect of the compatibility of WEM with FJ in SZD on malignant ascites and elucidate the potential mechanism from the perspective of the modulation of gut microbiota and related metabolic function. METHODS Qualitative and quantitative evaluation of main components was conducted for comprehensive characterization of SZD and WEM. The effect of WEM and SZD was compared on malignant ascites effusion (MAE) rats. The intestinal injury was evaluated by HE staining and oxidative damage. Ascites weight, urine amount, fecal water content, the expression of aquaporins, and cytokines in ascites (IL-6, VEGF, and TNF-α) were measured to estimate the water-expelling activity. The intestinal flora was detected by 16S rDNA sequencing and the content of fecal short-chain fatty acids (SCFAs) was analyzed using gas chromatography-mass spectrometry. Pseudo-germ-free (PGF) and fecal bacteria transplantation animal experiments were subsequently employed to validate this finding. The fecal metabolomics and correlation analysis were finally conducted to explore the related metabolic changes. RESULTS 51 and 33 components were identified in SZD and WEM, respectively. Compared to WEM alone, the compatibility with FJ remarkably reduced intestinal oxidative damage in MAE rats. Ascites was also relieved by downregulating the expression of AQP3 in the colon and decreasing the levels of IL-6, TNF-α and VEGF in ascites. The diversity of gut microbiota was reversed with an increase in Lactobacillus and Clostridia_UCG-014 while a decrease in Colidextribacter. Under the PGF condition, compatibility of WEM with FJ failed to reduce intestinal injury and alleviate MA significantly, but this effect was further enhanced after FMT. 23 potential fecal metabolites were finally identified. Correlation analysis further showed that Lactobacillus and Clostridia_UCG-014 were positively correlated with SCFAs and l-tryptophan. Colidextribacter was negatively correlated with thymidine but positively correlated with ursodeoxycholic acid and deoxycholic acid. CONCLUSION FJ cooperated with WEM reduced intestinal injury and alleviated malignant ascites by modulating gut microbiota, short-chain fatty and tryptophan metabolism. These findings provide a scientific basis for the clinical application of FJ from SZD and the safe usage of SZD.
Collapse
Affiliation(s)
- Shi-Kang Zhou
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Changzhou Key Laboratory of Human Use Experience Research & Transformation of Menghe Medical Sect, Changzhou Hospital of Chinese Medicinal Affiliated to Nanjing University of Chinese Medicine, No. 25 Heping North Road, Tianning District, Changzhou, 213003, PR China
| | - Jin-Di Xu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Xiao-Qin Gao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Ren-Jie Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Fang-Fang Cheng
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China
| | - Wei-Feng Yao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Taizhou Key Laboratory for Development of Traditional Chinese Medicine Health Products, Taizhou Engineering Research Center for Quality and Industrialization of Traditional Chinese Medicine, Nanjing University of Chinese Medicine Hanlin College, No.6, Kuangshi Road, Pharmaceutical High-tech District, Taizhou 225300, PR China
| | - Yi Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China.
| | - Ting Geng
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Taizhou Key Laboratory for Development of Traditional Chinese Medicine Health Products, Taizhou Engineering Research Center for Quality and Industrialization of Traditional Chinese Medicine, Nanjing University of Chinese Medicine Hanlin College, No.6, Kuangshi Road, Pharmaceutical High-tech District, Taizhou 225300, PR China.
| | - Li Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Qixia District, Nanjing 210023, PR China; Taizhou Key Laboratory for Development of Traditional Chinese Medicine Health Products, Taizhou Engineering Research Center for Quality and Industrialization of Traditional Chinese Medicine, Nanjing University of Chinese Medicine Hanlin College, No.6, Kuangshi Road, Pharmaceutical High-tech District, Taizhou 225300, PR China.
| |
Collapse
|
2
|
Liang Q, Hu JX, Liang ZS, Xiao LL, Xu WH. Euphorbia pekinensis Rupr. roots: a comprehensive review of botany, traditional uses, phytochemistry, pharmacology, quality control, and toxicology. Nat Prod Res 2024:1-17. [PMID: 39205630 DOI: 10.1080/14786419.2024.2397554] [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: 04/16/2024] [Revised: 07/23/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Euphorbia pekinensis Rupr. is a traditional herb generally distributed in most areas of China, north Korea and Japan. The dried roots of Euphorbia pekinensis Rupr. (REP), famous as 'Jing Da Ji' () have been applied as traditional herb medicines to expel water and rheum; disperse swelling, dissipate binds and to treat edoema, pleural effusions, uraemia, nephritis, cirrhosis with ascites, as well as other diseases. Recent advances in botany, traditional uses, phytochemistry, pharmacology, quality control, and toxicology of E. pekinensis roots are methodically outlined and current limitations as well as future perspectives also are discussed in order to guide scientifical investigation and rational application of REP. Up to now, 79 structurally diverse compounds have been obtained and characterised from REP, principally including diterpenoids, triterpenoids, tannins, phenols, and 29 volatile constituents. Among which, diterpenoids are considered as primary characteristic and active constituents. The extracts and individual compounds from REP have demonstrated significant pharmacological effects such as diuretic and purgative, anti-inflammatory, and cytotoxic effects. REP are widely used in traditional medicine due to diverse chemical constituents with obvious pharmacological effects. Modern phytochemical and pharmacological studies justified and explained relevant traditional uses of REP and offer worthy clues for new medical fields of industrial application. Nevertheless, a great number of thorough and detailed investigations should be carried out in active constituents, mechanisms of action, quality-marker, toxicology assessment, and detoxification mechanisms of REP.
Collapse
Affiliation(s)
- Qian Liang
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming, PR China
| | - Jia-Xin Hu
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming, PR China
| | - Zong-Suo Liang
- Shaoxing Academy of Biomedicine of Zhejiang Sci-Tech University, Shaoxing, PR China
| | - Li-Lin Xiao
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming, PR China
| | - Wen-Hui Xu
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming, PR China
- Shaoxing Academy of Biomedicine of Zhejiang Sci-Tech University, Shaoxing, PR China
| |
Collapse
|
3
|
Qian M, Ren X, Mao P, Li Z, Qian T, Wang L, Liu H. Transcriptomics-based analysis reveals the nephrotoxic effects of triphenyltin (TPT) on SD rats by affecting RAS, AQPs and lipid metabolism. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 199:105792. [PMID: 38458666 DOI: 10.1016/j.pestbp.2024.105792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 03/10/2024]
Abstract
Triphenyltin (TPT) is a class of organotin compounds that are extensively used in industry and agriculture. They have endocrine-disrupting effects and cause severe environmental contamination. Pollutants may accumulate in the kidneys and cause pathological complications. However, the mechanism of TPT's toxicological effects on the kidney remains unclear. This study aimed to investigate the toxic effects and mechanism of action of TPT exposure on renal impairment in rats. Male SD rats were divided into four groups: the Ctrl group (control group), TPT-L group (0.5 mg/kg/d), TPT-M group (1 mg/kg/d), and TPT-H group (2 mg/kg/d). After 28 days of exposure to TPT, we observed the morphology and structure of kidney tissue using HE, PASM, and Masson staining. We also detected serum biochemical indexes, performed transcriptome sequencing of rat kidney tissue using RNA-seq. Furthermore, protein expression levels were measured through immunohistochemistry and gene expression levels were determined using RT-qPCR. The study results indicated a decrease in kidney weight and relative kidney weight after 28 days of exposure to TPT. Additionally, TPT caused damage to kidney structure and function, as evidenced by HE staining, PASM staining, and serum biochemical tests. Transcriptomics identified 352 DEGs, and enrichment analyses revealed that TPT exposure primarily impacted the renin-angiotensin system (RAS). The expression levels of water channel proteins were reduced, and the expression levels of RAS and lipid metabolism-related genes (Mme, Ace, Fasn, Cyp4a8, Cpt1b and Ppard) were significantly decreased in the TPT-treated group. In summary, exposure to TPT may impair renal structure and function in rats by affecting RAS, AQPs, and lipid metabolism.
Collapse
Affiliation(s)
- Mingqing Qian
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Xijuan Ren
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Penghui Mao
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Zhi Li
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Tingting Qian
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China
| | - Li Wang
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China.
| | - Hui Liu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China.
| |
Collapse
|
4
|
Zulkifli MH, Abdullah ZL, Mohamed Yusof NIS, Mohd Fauzi F. In silico toxicity studies of traditional Chinese herbal medicine: A mini review. Curr Opin Struct Biol 2023; 80:102588. [PMID: 37028096 DOI: 10.1016/j.sbi.2023.102588] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/15/2023] [Accepted: 03/05/2023] [Indexed: 04/09/2023]
Abstract
With the availability of public databases that store compound-target/compound-toxicity information, and Traditional Chinese medicine (TCM) databases, in silico approaches are used in toxicity studies of TCM herbal medicine. Here, three in silico approaches for toxicity studies were reviewed, which include machine learning, network toxicology and molecular docking. For each method, its application and implementation e.g., single classifier vs. multiple classifier, single compound vs. multiple compounds, validation vs. screening, were explored. While these methods provide data-driven toxicity prediction that is validated in vitro and/or in vivo, it is still limited to single compound analysis. In addition, these methods are limited to several types of toxicity, with hepatotoxicity being the most dominant. Future studies involving the testing of combination of compounds on the front end i.e., to generate data for in silico modeling, and back end i.e., validate findings from prediction models will advance the in silico toxicity modeling of TCM compounds.
Collapse
Affiliation(s)
- Muhammad Harith Zulkifli
- Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Zafirah Liyana Abdullah
- Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | | | - Fazlin Mohd Fauzi
- Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Collaborative Drug Discovery Research, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
5
|
Song S, Qiu R, Jin X, Zhou Z, Yan J, Ou Q, Liu X, Li W, Mao Y, Yao W, Lu T. Mechanism exploration of ancient pharmaceutic processing (Paozhi) improving the gastroprotective efficacy of Aucklandiae Radix. JOURNAL OF ETHNOPHARMACOLOGY 2022; 287:114911. [PMID: 34902533 DOI: 10.1016/j.jep.2021.114911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Processing, also called Paozhi in Chinese, is an ancient Chinese pharmaceutic processing technique developed along with the Chinese herbal medicines (CHMs). The understanding of the mechanism of Paozhi has been investigated for several decades. Aucklandiae Radix (CAR) and its roasted processed products are all used in indigestion as a kind of CHMs. Processed Aucklandiae Radix (PAR) had a stronger effect to protect gastric mucosa than CAR, while the main compounds in CAR were reduced sharply after being processed. The underlying mechanism of this phenomenon is still unclear. AIM OF THE STUDY This study was aimed to evaluate whether PAR have a stronger gastroprotective effect than CAR and the underlying mechanisms of such circumstance. MATERIALS AND METHODS Ultra-fast liquid chromatography coupled with quadrupole time of flight mass spectrometry (UFLC-QTOF-MS/MS) coupled with multivariate statistical analyses was employed to explore chemical compounds which had a relatively stable content in PAR. Based on the compounds selected as the research object, network pharmacology was applied to visualize the relationships between the selected components and the gastroprotective-related targets from disease database, at the same time the possible intervention path of CAR/PAR which might be responsible for the effect of CAR/PAR on gastritis-induced rats was also built. Then, the key proteins were detected by western blotting to verify and compare the pharmacological effects of CAR/PAR. RESULTS Through UFLC-QTOF-MS/MS and orthogonal partial least squares discriminant analysis (OPLS-DA), sixteen compounds stable in PAR were discovered, of which saussureamine C and saussureamine B were estimated as the core compounds to exert gastroprotective in PAR predicted by network pharmacology analysis. Under the guide of KEGG pathway enrichment analysis, PI3K/AKT, p38 MAPK (Mitogen-activated protein kinase) and nuclear factor-kappa B (NF-κB) signaling pathways were forecasted as the possible healing mechanisms of CAR/PAR, and that result was verified by the experiments in vivo. PAR performed a stronger ability to reduce the level of p38 MAPK and NF-κB p65 than CAR, which may partially explain the different ability of CAR/PAR against gastric mucosa damage. CONCLUSION This study clarified that although Paozhi entailed a sharp decrease on the main compounds of CAR, there were some compounds which were not sensitive to high temperature and preserved in PAR and had a relative higher content in PAR than in CAR. PAR has stronger influence on MAPKs/NF-κB signaling pathway than CAR, which may reveal that the stronger gastroprotective effect of PAR perhaps rely on the constitutions with a higher relative abundance after Paozhi. The present research combined UFLC-QTOF-MS/MS and network pharmacology deeply investigated the impact of the roasted processing on the chemical constitutions and gastroprotective effect of CAR and offered reference for the clinical application of CAR/PAR.
Collapse
Affiliation(s)
- Shen Song
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Rongli Qiu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaodan Jin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zhuxiu Zhou
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jin Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Qiaochan Ou
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiao Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Weidong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yiqing Mao
- Nanjing Institute for Food and Drug Control, Nanjing, 211198, China.
| | - Weifeng Yao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Tulin Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
6
|
The Mechanism of Ginseng and Astragalus Decoction in the Treatment of Malignant Pleural Effusion Based on Network Pharmacology and Molecular Docking Technology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7731402. [PMID: 35341150 PMCID: PMC8941536 DOI: 10.1155/2022/7731402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/31/2022] [Indexed: 11/17/2022]
Abstract
Introduction The objective of our study is to explore the potential active ingredients and activity of Ginseng and Astragalus decoction (GAD) in the treatment of malignant pleural effusion (MPE) by using network pharmacology and molecular docking technologies. Methods The active ingredients and corresponding targets of Ginseng and Astragalus were extracted from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform. The relevant targets of malignant pleural effusion (MPE) were searched in the disease databases. Overlapping targets of Ginseng and Astragalus and the corresponding targets of MPE were obtained to define the effective target of GAD for the treatment of MPE. The STRING database was applied to construct a predicted protein-protein interaction network for intersected targets. The Cytoscape software was used to screen key targets with a therapeutic potential. Using the Metascape database, we performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis on the targets identified in the study. PyMOL and AutoDock Vina were used to molecularly dock the selected key components to their respective key targets for MPE treatment. Results The core target network revealed 22 main active ingredients, 26 main targets, and 16 signaling pathways in GAD. Molecular docking revealed 6 targets (AKT serine/threonine kinase 1, intercellular adhesion molecule, Jun proto-oncogene, peroxisome proliferator activated receptor gamma, prostaglandin-endoperoxide synthase 2, and tumor necrosis factor) that could partially dock with kaempferol, frutinone A, ginsenoside RH2, formononetin, and quercetin. Conclusions Several components, targets, and signaling pathways of GAD contribute to the treatment of MPE, which suggests a rationale for further investigation on GAD's active molecule and mechanism of action in the clinical application of MPE.
Collapse
|
7
|
Bailly C. Yuanhuacin and Related Anti-Inflammatory and Anticancer Daphnane Diterpenes from Genkwa Flos-An Overview. Biomolecules 2022; 12:192. [PMID: 35204693 PMCID: PMC8961543 DOI: 10.3390/biom12020192] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
The dried flower buds of the plant Daphne genkwa Sieb. et Zucc. have been largely used in traditional Chinese medicine for the treatment of inflammatory diseases. Numerous diterpenoids have been isolated from the Genkwa Flos (yuanhua in Chinese), including a series of daphnane-type diterpene designated as yuanhuacin (YC, often improperly designated as yuanhuacine) and analogues with a patronymic name. The series includes ten daphnane-type diterpenes: yuanhuacin, yuanhuadin (YD), yuanhuafin (YF), yuanhuagin (YG), yuanhuahin (YH), yuanhuajin (YJ), yuanhualin (YL), yuanhuamin (YM), yuanhuapin (YP), and yuanhuatin (YT). They are distinct from the rare flavonoid yuanhuanin. The series comprises several anticancer agents, such as the lead compound YC, which has revealed potent activity in vitro and in vivo against models of lung and breast cancers. The main signaling pathways implicated in the antitumor effects have been delineated. Protein kinase C is a key factor of activity for YC, but in general the molecular targets at the origin of the activity of these compounds remain little defined. Promising anticancer effects have been reported with analogues YD and YT, whereas compounds YF and YP are considered more toxic. The pharmacological activity of each compound is presented, as well as the properties of Genkwa Flos extracts. The potential toxic effects associated with the use of these compounds are also underlined.
Collapse
|
8
|
He J, Yang L. Diuretic effect of Lagopsis supina fraction in saline-loaded rats is mediated through inhibition of aquaporin and renin-angiotensin-aldosterone systems and up-regulation of atriopeptin. Biomed Pharmacother 2021; 139:111554. [PMID: 33845373 DOI: 10.1016/j.biopha.2021.111554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 12/31/2022] Open
Abstract
Lagopsis supina (Steph. ex Willd.) lk. -Gal. ex Knorr. has been used as a diuretic agent in China for centuries with limited scientific evidence. This study investigated the diuretic efficacy and underlying mechanism of a macroporous adsorption resin with 30% ethanol elution fraction from L. supina (LSC) in saline-loaded rats and to identify its phytochemicals by ultra-high-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (UHPLC-qTOF-MS/MS). As a result, 18 phenylpropanoids, 14 flavonoids and 15 others were identified in LSC, among which stachysoside A and acteoside could be the main bio-active constituents responsible for the diuretic effect. In parallel, the daily administration of LSC (16, 32 and 64 mg/kg) markedly promoted urinary excretion after 2 h of treatment. Moreover, LSC had no effect on urinary Na+ and K+ concentrations, as well as on serum Na+-K+-ATPase activity. Meanwhile, LSC significantly decreased the serum levels of angiotensin II (Ang II), anti-diuretic hormone (ADH), aldosterone (ALD), aquaporin (AQP) 1, AQP2 and AQP3, suppressed renal AQP1, AQP2, and AQP3 mRNA expressions, down-regulated AQP1, AQP2 and AQP3 protein levels, and up-regulated serum atriopeptin (ANP) level in a dose-dependent manner. These findings suggest that LSC has acute and prolonged diuretic effects by inhibiting the AQPs, RAAS, and upregulation of atriopeptin in saline-loaded rats, and this finding support LSC as a novel diuretic agent.
Collapse
Affiliation(s)
- Junwei He
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Li Yang
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|