1
|
Luo C, Tian L, Wen Y, Zheng Z. Protective Effects of Schizochytrium Microalgal Fatty Acids on Alcoholic Liver Disease: A Network Pharmacology and In Vivo Study. Assay Drug Dev Technol 2025. [PMID: 39815972 DOI: 10.1089/adt.2024.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025] Open
Abstract
This study aimed to elucidate the hepatoprotective mechanisms of microalgal fatty acids (MFA) from Schizochytrium against alcoholic liver disease (ALD) through network pharmacology and in vivo analysis. Network pharmacology and molecular docking methodologies were employed to predict the potential mechanisms of MFA against ALD. To substantiate these predictions, an acute alcoholic liver injury mouse model was utilized to assess the impact of MFA on serum levels of alanine aminotransferase (ALT), alkaline phosphatase (ALP), aspartate aminotransferase (AST), total protein (TP), and albumin (ALB). Additionally, liver histopathology and the expression levels of phosphatidylinositol 3 kinase (PI3K) and protein kinase B (AKT) protein were evaluated. Seven active ingredients and 53 potential targets (including 7 core targets) for ALD treatment were identified in MFA. Kyoto Encyclopedia of Genes and Genomes pathway analyses indicated that these seven core targets are implicated in various biological pathways, notably those associated with cancer, viral infections, and the PI3K/AKT signaling pathway. Furthermore, molecular docking studies demonstrated that docosahexaenoic acid and docosapentaenoic acid in MFA exhibited strong binding affinity for these seven crucial targets. Animal experiments demonstrated that administration of MFA significantly decreased the levels of AST, ALT, and ALP, while increasing the levels of ALB and TP in mice with acute alcoholic liver injury. Moreover, MFA ameliorated liver tissue pathology and markedly down-regulated the expression of PI3K and AKT proteins in the liver. These results suggest that MFA may possess therapeutic potential for ALD by targeting multiple pathways, with its mechanisms likely involving the inhibition of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Cailin Luo
- Department of Basic Medical Science, Quanzhou Medical College, Quanzhou, China
| | - Li Tian
- Department of Basic Medical Science, Quanzhou Medical College, Quanzhou, China
| | - Yangmin Wen
- Department of Basic Medical Science, Quanzhou Medical College, Quanzhou, China
| | - Zhihua Zheng
- Department of Basic Medical Science, Quanzhou Medical College, Quanzhou, China
| |
Collapse
|
2
|
Xu Q, Su W, Huang C, Zhong H, Huo L, Cai J, Li P. Multifunctional Polysaccharide Self-Healing Wound Dressing: NIR-Responsive Carboxymethyl Chitosan / Quercetin Hydrogel. Adv Healthc Mater 2024:e2403267. [PMID: 39551981 DOI: 10.1002/adhm.202403267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/29/2024] [Indexed: 11/19/2024]
Abstract
As the misuse of antibiotics increases bacterial resistance, the treatment of infected wounds caused by bacteria encounters significant challenges. Conventional antimicrobial dressings often fall short in their ability to inhibit bacterial infections while simultaneously promoting wound healing. To address this issue, a polysaccharide self-healing hydrogel (CPP@PDA/Que3) wound dressing is successfully developed by incorporating quercetin and polydopamine nanoparticles into a carboxymethyl chitosan matrix. The dressing can be easily injected locally to create a protective barrier over the wound, effectively stopping bleeding and rapidly inhibiting inflammation. Furthermore, the CPP@PDA/Que3 hydrogel exhibits remarkable antioxidant and antibacterial properties, stemming from the combination of quercetin and near-infrared (NIR) photothermal therapy. It demonstrates the ability to eliminate 99.52% of Staphylococcus aureus and 99.39% of Escherichia coli in in vitro antibacterial experiments. Additionally, the in vivo wound healing experiment shows a healing rate of ≈97%. The experimental results indicate that under NIR laser (808 nm) irradiation, the polysaccharide-based hydrogel dressing significantly inhibits bacterial growth, reduces oxidative stress, expedites angiogenesis, and thereby accelerates the transition from inflammation to wound healing. In summary, the CPP@PDA/Que3 hydrogel exhibits significant potential as a wound dressing, providing a novel approach for clinically advancing the treatment of bacterial wounds.
Collapse
Affiliation(s)
- Qiuting Xu
- Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Wei Su
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning, 530001, China
| | - Cuilan Huang
- Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Haiyi Zhong
- Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Lini Huo
- Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Jinyun Cai
- Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Peiyuan Li
- Guangxi University of Chinese Medicine, Nanning, 530001, China
| |
Collapse
|
3
|
Liu G, Zhang S, Lin R, Cao X, Yuan L. Anti-tumor target screening of sea cucumber saponin Frondoside A: a bioinformatics and molecular docking analysis. Front Oncol 2023; 13:1307838. [PMID: 38144520 PMCID: PMC10739435 DOI: 10.3389/fonc.2023.1307838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Cancer remains the leading cause of death worldwide. In spite of significant advances in targeted and immunotherapeutic approaches, clinical outcomes for cancer remain poor. The aim of the present study was to investigate the potential mechanisms and therapeutic targets of Frondoside A for the treatment of liver, pancreatic, and bladder cancers. The data presented in our study demonstrated that Frondoside A reduced the viability and migration of HepG2, Panc02, and UM-UC-3 cancer cell in vitro. Moreover, we utilized the GEO database to screen and identify for differentially expressed genes (DEGs) in liver, pancreatic, and bladder cancers, which resulted in the identification of 714, 357, and 101 DEGs, respectively. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation were performed using the Metascape database for DEGs that were significantly associated with cancer development. The protein-protein interaction (PPI) networks of the identified DEGs in liver, pancreatic, and bladder cancers were analyzed using Cytoscape 3.9.0 software, and subsequently identified potential key genes that were associated with these networks. Subsequently, their prognostic values were assessed by gene expression level analysis and Kaplan-Meier survival analysis (GEPIA). Furthermore, we utilized TIMER 2.0 to investigate the correlation between the expression of the identified key gene and cancer immune infiltration. Finally, molecular docking simulations were performed to assess the affinity of Frondoside A and key genes. Our results showed a significant correlation between these DEGs and cancer progression. Combined, these analyses revealed that Frondoside A involves in the regulation of multiple pathways, such as drug metabolism, cell cycle in liver cancer by inhibiting the expression of CDK1, TOP2A, CDC20, and KIF20A, and regulates protein digestion and absorption, receptor interaction in pancreatic cancer by down-regulation of ASPM, TOP2A, DLGAP5, TPX2, KIF23, MELK, LAMA3, and ANLN. While in bladder cancer, Frondoside A regulates muscle contraction, complement and coagulation cascade by increase FLNC expression. In conclusion, the present study offers valuable insights into the molecular mechanism underlying the anticancer effects of Frondoside A, and suggests that Frondoside A can be used as a functional food supplement or further developed as a natural anti-cancer drug.
Collapse
Affiliation(s)
- Guangchun Liu
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shenglin Zhang
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ruoyan Lin
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xudong Cao
- Deparment of Chemical and Biological Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Lihong Yuan
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
4
|
Wang Y, Li X, Qi M, Li X, Zhang F, Wang Y, Wu J, Shu L, Fan S, Li Y, Li Y. Pharmacological effects and mechanisms of YiYiFuZi powder in chronic heart disease revealed by metabolomics and network pharmacology. Front Mol Biosci 2023; 10:1203208. [PMID: 37426419 PMCID: PMC10327484 DOI: 10.3389/fmolb.2023.1203208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: YiYiFuZi powder (YYFZ) is a classical formula in Chinese medicine, which is commonly used clinically for the treatment of Chronic Heart Disease (CHD), but it's pharmacological effects and mechanism of action are currently unclear. Methods: An adriamycin-induced CHD model rat was established to evaluate the pharmacological effects of YYFZ on CHD by the results of inflammatory factor level, histopathology and echocardiography. Metabolomic studies were performed on rat plasma using UPLC-Q-TOF/MS to screen biomarkers and enrich metabolic pathways; network pharmacology analysis was also performed to obtain the potential targets and pathways of YYFZ for the treatment of CHD. Results: The results showed that YYFZ significantly reduced the levels of TNF-α and BNP in the serum of rats, alleviated the disorder of cardiomyocyte arrangement and inflammatory cell infiltration, and improved the cardiac function of rats with CHD. The metabolomic analysis identified a total of 19 metabolites, related to amino acid metabolism, fatty acid metabolism, and other metabolic pathways. Network pharmacology showed that YYFZ acts through PI3K/Akt signaling pathway, MAPK signaling pathway and Ras signaling pathway. Discussion: YYFZ treatment of CHD modulates blood metabolic pattern and several protein phosphorylation cascades but importance specific changes for therapeutic effect require further studies.
Collapse
Affiliation(s)
- Yuming Wang
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xue Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Qi
- TIPRHUYA Advancing Innovative Medicines Ltd., Tianjin, China
| | - Xiaokai Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fangfang Zhang
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuyu Wang
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junke Wu
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lexin Shu
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Simiao Fan
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunfei Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yubo Li
- School of Chinese Materia, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
5
|
Gao X, Zhang Y, Li T, Li J, Su Y, Wang H, Yan Z, Qin K. Uncovering the molecular mechanisms of Fructus Choerospondiatis against coronary heart disease using network pharmacology analysis and experimental pharmacology. Anal Biochem 2023:115214. [PMID: 37353066 DOI: 10.1016/j.ab.2023.115214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/29/2023] [Accepted: 06/11/2023] [Indexed: 06/25/2023]
Abstract
Fructus Choerospondiatis (FC), a Mongolian medicine, was mainly used in Mongolian medical theory for the treatment of coronary heart disease (CHD). Nonetheless, the main components and mechanisms of action of FC in the treatment of coronary artery disease have not been studied clearly. AIM OF THE STUDY The aim of this study is to identify the components of FC and analyze the pathways affected by the targets of these components to probe into the potential mechanisms of action of FC on coronary heart disease. MATERIALS AND METHODS Identification of compounds in FC employing high performance liquid chromatography quadrupole time-of-flight tandem mass spectrometry (HPLC-QTOF-MS) method, then further investigate the network pharmacology and molecular docking to obtain potential targets and elucidate the potential mechanism of action of FC in the therapy of CHD. Experimental validation was established to verify the mechanism of FC in vitro. RESULTS 21 FC components were identified and 65 overlapping targets were gained. In addition, these ingredients regulated AMPK and PPAR signaling pathway by 65 target genes including IL6, AKT1 and PPARg, etc. Molecular docking displayed that the binding ability of the key target PPARg to FC components turned out to be better. Experimental validation proved that FC treatment decreased the expression of PPARg (p < 0.05) compare with model group, which may be involved in the PPAR signaling pathway. CONCLUSIONS This study was the first to elucidate the mechanism of action of components of FC for the treatment of CHD using network pharmacology. It alleviated CHD by inhibiting the expression of PPARg to attenuate hypoxia/reoxygenation injury, and the results give a basis for elucidating the molecular mechanism of action of FC for the treatment of coronary heart disease.
Collapse
Affiliation(s)
- Xun Gao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222001, China; School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Yue Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, 222005, China; Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222001, China; School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Tingting Li
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222001, China; School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jioajiao Li
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222001, China; School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yingying Su
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222001, China; School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Hongsen Wang
- Jiangsu Yuanchuang Pharmaceutical Research and Development Co., Ltd, China
| | - Zhankuan Yan
- Jiangsu Yuanchuang Pharmaceutical Research and Development Co., Ltd, China
| | - Kunming Qin
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222001, China; School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
6
|
Li YL, Qin SY, Li Q, Song SJ, Xiao W, Yao GD. Jinzhen Oral Liquid alleviates lipopolysaccharide-induced acute lung injury through modulating TLR4/MyD88/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154744. [PMID: 36934667 DOI: 10.1016/j.phymed.2023.154744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/12/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Acute lung injury (ALI) has the attribution of excessive inflammation of the lung. Jinzhen oral liquid (JO), a famous Chinese recipe used to treat ALI, has a favorable therapeutic effect on ALI. However, its anti-inflammatory mechanism has not been extensively studied. PURPOSE This study was to elucidate the effects of JO on lipopolysaccharide (LPS)-induced ALI and its molecular mechanism. METHODS An ALI model was established by intratracheal instillation of LPS (2 mg/50 μl). The open field experiment was carried out to explore the spontaneous movement and exploratory behavior of ALI mice. Cytokines levels concentrations (IL-6, IL-10 and TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). Network pharmacology was used to predict the mechanism of JO against ALI. Immunofluorescence, co-immunoprecipitation, fluorescence resonance energy transfer (FRET), Western blot and RT-PCR were used to verify the molecular mechanisms of JO. RESULTS The in vivo results suggested that JO (1, 2, 4 g/kg) dose-dependently improved the exercise performance of mice and reduced the lung W/D weight ratio as well as the production of IL-6 and TNF-α, but increased the release of IL-10 in the ALI group. The network pharmacological analysis demonstrated that the Toll-like receptor (TLR) pathway might be the fundamental action mechanisms of JO against ALI. Immunofluorescence staining and co-immunoprecipitation analysis showed that JO decreased the expression levels of TLR4 and MyD88 and reduced their interaction in the lung tissue of ALI mice. Meanwhile, JO decreased nuclear translocation and phosphorylation of NF-κB P65. The results from cellular experiments were in line with those in vivo. The FRET experiment also confirmed that JO disturbed the interaction of TLR4 and MyD88. Subsequently, we also found that the six indicative components of JO have the similar therapeutic effect as JO. CONCLUSIONS In summary, we suggested that JO suppressed the TLR4/MyD88/NF-κB signaling pathway, thus inhibiting LPS-induced ALI in vitro and in vivo. The clarified mechanism provided an important theoretical basis and a novel treatment strategy for the ALI treatment of JO.
Collapse
Affiliation(s)
- Ya-Ling Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shu-Yan Qin
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qian Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, China.
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, China.
| |
Collapse
|
7
|
Li X, Gong YX, Feng L, Wang XJ, Wang JW, Zhang AX, Tan NH, Wang Z. Neuropyrones A-E, five undescribed α-pyrone derivatives with tyrosinase inhibitory activity from the endophytic fungus Neurospora dictyophora WZ-497. PHYTOCHEMISTRY 2023; 207:113579. [PMID: 36586529 DOI: 10.1016/j.phytochem.2022.113579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Five undescribed α-pyrone derivatives, named neuropyrones A-E, were isolated from the endophytic fungus Neurospora dictyophora WZ-497 derived from the stems of Aster tataricus L. f. The structures of these α-pyrones with absolute configurations were determined by comprehensive spectroscopic analysis and computational calculations. All isolated compounds were tested for various bioactivities, including tyrosinase inhibitory activity. The results showed that neuropyrones A-C displayed potent inhibitory effects on tyrosinase with IC50 values of 0.38 ± 0.07, 0.49 ± 0.06, and 0.12 ± 0.01 mM, respectively, which were comparable to that of the positive control, kojic acid (IC50 = 0.14 ± 0.021 mM). A molecular docking study revealed the interaction between 3 and the His263, His85, Val283, Asn260, Phe264, and Val248 residues of tyrosinase.
Collapse
Affiliation(s)
- Xin Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuan-Xiang Gong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Li Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin-Jia Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jing-Wen Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - An-Xin Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ning-Hua Tan
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Zhe Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
8
|
Zhang Z, Wang X, Zhang X, Wu J, Chen J, Li W. Integrated LC-MS and network pharmacology methods to screen quantitative indicators in the Hippocampus histrix Kaup and method transfer. J Pharm Biomed Anal 2023; 228:115294. [PMID: 36827860 DOI: 10.1016/j.jpba.2023.115294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/10/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Hippocampus histrix Kaup is a popular marine medicine with high medicinal and healthcare values. In this study, liquid chromatography-mass spectrometry (LC-MS) analysis combined with network pharmacological method was used to screen for suitable quantitative indicators for the quality control of H. histrix Kaup. Firstly, an LC-MS analytical method for the simultaneous determination of 12 nucleosides in extracts of H. histrix Kaup was established. And then, a network pharmacological method incorporated target prediction, protein-protein interaction network, components-targets network, and targets-pathways network was performed to screen for quantitative indicators. Finally, the developed LC-MS method was transferred to liquid chromatographs to improve the generalizability of the method. All 12 nucleotides were authenticated in extracts of H. histrix Kaup by comparing with the standards. The optimal chromatographic separation conditions are as follows: the chromatographic separation was achieved on an Acquire BEH-C18 column (2.1 mm * 100 mm, 1.7 µm) and gradient elution was performed using methanol solution and buffer (0.30% formic acid and 10 mmol/L ammonium acetate) as mobile phase at a flow rate of 0.15 mL/min and an acquisition wavelength of 260 nm. Network pharmacology results showed that adenosine, and uridine show excellent pharmacological activity. Integration the content, correlation, chromatographic separation, and pharmacological activity of each compound in H. histrix Kaup, uridine and adenosine were tentatively determined as quantitative indicators for quality control in H. histrix Kaup. The established LC-MS method was successfully transferred to liquid chromatographs, and the method is stable and reliable for the quality control of H. histrix Kaup. This developed integrated strategy was successfully used to screen quantitative indicators in the H. histrix Kaup.
Collapse
Affiliation(s)
- Zhiyong Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xi Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoyang Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiaheng Wu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Junhui Chen
- Marine Bioresource and Environment Research Center, Key Laboratory of Marine Eco-Environmental Science and Technology, The First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Qingdao Key Lab on Analytical Technology Development and Standardization of Chinese Medicines, The First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China.
| | - Wenlong Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
9
|
Wang X, Zhang C, Liu L, Zhong Y, Wang Y, Liu F, Zhu J, Mu Z, Zhang S, Wang X, Zhong G, Liang J, Zeng J. Effective materials and mechanisms study of Tibetan herbal medicine Lagotis integra W. W. Smith treating DSS-induced ulcerative colitis based on network pharmacology, molecular docking and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115800. [PMID: 36228890 DOI: 10.1016/j.jep.2022.115800] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/13/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lagotis integra W. W. Smith (L. integra W. W. Smith) is an important origin plant of the famous Tibetan medicine HERBA LAGOTIS. It was documented to treat "Chi Ba" disease clinically, the symptoms of which are similar to ulcerative colitis (UC). AIMS OF THIS STUDY To screen out the active components and study the mechanisms of L. integra W. W. Smith treating UC. MATERIALS AND METHODS The components of L. integra W. W. Smith were comprehensively analyzed using UHPLC-Q-TOF/MS method. The mechanisms were investigated using network pharmacology method including target prediction, protein-protein interaction network analysis and gene enrichment analysis. Then, the mechanisms were verified using Dextran Sulfate Sodium (DSS)-induced UC model. Finally, the core active components were further screened out through molecular docking. RESULTS The results showed that 32 major components were identified including 8 flavonoids, 9 phenylpropanoid glycosides, 13 iridoid glycosides and 1 phenolic acid. 76 potential core therapeutic targets and top 5 key targets, which were AKT serine/threonine kinase 1 (AKT1), vascular endothelial growth factor (VEGFA), tumor necrosis factor-α (TNF-α), epidermal growth factor receptor (EGFR) and caspase-3 (CASP3), were screened out according to network pharmacology analysis. Animal experiments confirmed that those compounds could downregulate the expression levels of the 5 key target proteins in colonic tissue of mice to exert excellent anti-UC effect. Molecular docking results showed that the main active components were echinacoside, hemiphroside B, plantamajoside, plantainoside D, 10-O-trans-isoferuloyl catalpol and scutellarioside II. CONCLUSIONS For the first time, our study provides insights into the effective materials and molecular mechanisms of L. integra W. W. Smith treating UC, which contributes to the understanding of its pharmacodynamics.
Collapse
Affiliation(s)
- Xinhong Wang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Chi Zhang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Lin Liu
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yuanhan Zhong
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yujie Wang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Fangyuan Liu
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jixiao Zhu
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Zejing Mu
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shouwen Zhang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Xiaomin Wang
- School of Chinese Medicines and Life Science, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Guoyue Zhong
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jian Liang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Jinxiang Zeng
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| |
Collapse
|
10
|
Ai YF, Dong SH, Lin B, Huang XX, Song SJ. Acylated sucroses and butenolide analog from the leaves of Tripterygium wilfordii Hook. f. and their potential anti-tyrosinase effects. Fitoterapia 2022; 161:105250. [DOI: 10.1016/j.fitote.2022.105250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 11/04/2022]
|
11
|
Chemical structures and anti-tyrosinase activity of the constituents from Elephantopus scaber L. Fitoterapia 2022; 162:105259. [PMID: 35931288 DOI: 10.1016/j.fitote.2022.105259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 11/24/2022]
Abstract
Four undescribed compounds including one germacrane-type sesquiterpene lactones (1), alkaloid (2) along with two neolignans (3-4) were isolated from Elephantopus scaber L. Their structures and absolute configurations were elucidated unambiguously by means of 1D and 2D NMR spectroscopic data analysis, and quantum chemical electronic circular dichroism calculations, as well as single-crystal X-ray crystallography. Their anti-tyrosinase activities have been evaluated in vitro and compound 2 exhibited significant inhibitory activity. Furthermore, molecular docking was performed to study the interaction patterns between 2 and the tyrosinase.
Collapse
|
12
|
Qian Z, Yu J, Chen X, Kang Y, Ren Y, Liu Q, Lu J, Zhao Q, Cai M. De Novo Production of Plant 4'-Deoxyflavones Baicalein and Oroxylin A from Ethanol in Crabtree-Negative Yeast. ACS Synth Biol 2022; 11:1600-1612. [PMID: 35389625 DOI: 10.1021/acssynbio.2c00026] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Baicalein and oroxylin A are well-known medicinal 4'-deoxyflavones found mainly in the roots of traditional medicinal plant Scutellaria baicalensis Georgi. However, extraction from plants is time-consuming, environmentally unfriendly, and insufficient. Although microbial synthesis of flavonoids has been extensively reported, synthesis of downstream modified 4'-deoxyflavones has not, and their yields are extremely low. Here, we reassembled the S. baicalensis 4'-deoxyflavone biosynthetic pathway in a Crabtree-negative yeast, Pichia pastoris, with activity analysis and combinatorial expression of eight biosynthetic genes, allowing production of 4'-deoxyflavones like baicalein, oroxylin A, wogonin, norwogonin, 6-methoxywogonin, and the novel 6-methoxynorwogonin. De novo baicalein synthesis was then achieved by complete pathway assembly. Toxic intermediates highly impaired the cell production capacity; hence, we alleviated cinnamic acid growth inhibition by culturing the cells at near-neutral pH and using alcoholic carbon sources. To achieve pathway balance and improve baicalein and oroxylin A synthesis, we further divided the pathway into five modules. A series of ethanol-induced and constitutive transcriptional amplification devices were constructed to adapt to the modules. This fine-tuning pathway control considerably reduced byproduct and intermediate accumulation and achieved high-level de novo baicalein (401.9 mg/L with a total increase of 1182-fold, the highest titer reported) and oroxylin A (339.5 mg/L, for the first time) production from ethanol. This study provides new strategies for the microbial synthesis of 4'-deoxyflavones and other flavonoids.
Collapse
Affiliation(s)
- Zhilan Qian
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiahui Yu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xinjie Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yijia Kang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yanna Ren
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jian Lu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qing Zhao
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai 201602, China
- State Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Menghao Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
13
|
Liu H, Xu Y, Wang Y, Liu C, Chen J, Fan S, Xie L, Dong Y, Chen S, Zhou W, Li Y. Study on endocrine disruption effect of paclobutrazol and uniconazole on the thyroid of male and female rats based on lipidomics. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113386. [PMID: 35286959 DOI: 10.1016/j.ecoenv.2022.113386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
The present study investigated the effects of paclobutrazol and uniconazole on thyroid endocrine system in rats. Lipidomic analysis was performed to obtain the biomarkers of thyroid endocrine disruption induced by paclobutrazol and uniconazole. Network pharmacology was further used to discover potential targets of biomarkers related to drugs and diseases. After paclobutrazol and uniconazole administration, seven and four common biomarkers related to thyroid endocrine disruption for female and male rats were obtained, respectively. Paclobutrazol and uniconazole significantly increased the biomarker levels of PG (12:0/15:0), PS (14:0/16:0), PA (20:1/15:0) and PG (13:0/17:0) in both sexes of rats. Exposure to paclobutrazol additionally caused a significant decrease of PG (22:6/20:2), PE (24:1/18:1) and PE (24:0/18:0) in female rats, while an increase in male rats. Changes of the common biomarkers for paclobutrazol and uniconazole revealed similar endocrine disruption effect, which was higher in the females. Network pharmacology and KEGG pathway analysis indicated that the thyroid endocrine disrupting effects of paclobutrazol and uniconazole may be related to V-akt murine thymoma viral oncogene homolog (Akts), mitogen-activated protein kinase (MAPKs), epidermal growth factor receptor (EGFR), Insulin-like growth factor (IGF-1), IGF-IR and V-Raf murine sarcoma viral oncogene homolog B1 (BRAF). The results demonstrated that paclobutrazol and uniconazole could cause thyroid endocrine disorders in male and female rats, which were sex-specific, thus highlighting the importance of safe and effective application of these plant growth regulators.
Collapse
Affiliation(s)
- Hui Liu
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Yanyan Xu
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Yuming Wang
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Chunyang Liu
- National Aquatic Germplasm Resources Conservation Area Management Office in Qinhuangdao, Daihe Fishing Port, West Beach Road, Beidaihe District, Qinhuangdao, Hebei 066000, China
| | - Jun Chen
- Animal Health Supervision Office in Qinhuangdao, No. 52, Guancheng South Road, Shanhaiguan District, Qinhuangdao, Hebei 066000, China
| | - Simiao Fan
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Lijuan Xie
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Yaqian Dong
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Siyu Chen
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Wenjie Zhou
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Yubo Li
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, West Zone, Tuanbo New City, Jinghai District, Tianjin 301617, China.
| |
Collapse
|