1
|
Zhang W, Cai S, Luan W, Ding M, Di L. Integrated serum pharmacochemistry, network pharmacology and experimental verification to explore the mechanism of Aconiti Lateralis Radix Praeparata in treatment of lung cancer. J Pharm Biomed Anal 2025; 252:116472. [PMID: 39278160 DOI: 10.1016/j.jpba.2024.116472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Aconiti Lateralis Radix Praeparata (Fuzi) is a traditional Chinese medicine (TCM) widely used in treating cancer. Our formerly investigations confirmed the anti-lung cancer efficacy of Fuzi, but systematic analysis of the ingredients of Fuzi absorbed into serum and the corresponding molecular mechanism in treating lung cancer remained unknown. In this work, UPLC-Q-TOF-MS was applied to detect the ingredients of Fuzi in rat serum. Next, the possible targets and key pathways of the components absorbed into serum of Fuzi were predicted by network pharmacology. Then, the binding activity of components and potential targets were performed by molecular docking. Afterwards, the proliferation, mitochondrial membrane potential (MMP), apoptosis and reactive oxygen species (ROS) of lung cancer cells after treatment with Fuzi-containing serum were determined by MTT assay, JC-1 fluorescent probe, Annexin V-FITC/PI double staining and DCFH-DA respectively. Finally, the predicted target was further validated with qRT-PCR. In total, identification of 20 components of Fuzi derived from rat serum were achieved. The prediction of network pharmacology indicated that these compounds might exert their therapeutic effects by modulating mTOR. The findings from molecular docking proved that fuziline, songorine, napelline and hypaconitine exhibited binding potential with the mTOR. Cancer cell experiments revealed that the Fuzi-containing serum inhibited cell proliferation, induced apoptosis, reduced MMP and increased ROS. Additionally, Fuzi-containing serum significantly reduced the mRNA expression of mTOR. This study revealed that fuziline, songorine, napelline and hypaconitine were the main ingredients of Fuzi absorbed into serum. Furthermore, Fuzi-containing serum demonstrated inhibitory effects on the proliferation of lung cancer cells and induced the apoptosis. Combined with the results of network pharmacology, molecular docking and biological verification, Fuzi-containing serum might exert its anti-lung cancer effect by inhibiting mTOR. This study would provide a deeper understanding of Fuzi in treating lung cancer and offer a scientific reference for its clinical utilization.
Collapse
Affiliation(s)
- Wen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of Traditional Chinese Medicine, Nanjing 210023, China.
| | - Shuhui Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of Traditional Chinese Medicine, Nanjing 210023, China
| | - Wenhao Luan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of Traditional Chinese Medicine, Nanjing 210023, China
| | - Menglei Ding
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of Traditional Chinese Medicine, Nanjing 210023, China; Department of Pharmacy, Kunshan Hospital of Chinese Medicine, Suzhou China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of Traditional Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Li LY, Liu SZ, Yu X, Shi X, You H, Liu P, Wang F, Wang P, Chen LL. Liuwei Anshen Capsule alleviates cognitive impairment induced by sleep deprivation by reducing neuroapoptosis and inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 341:119311. [PMID: 39743184 DOI: 10.1016/j.jep.2024.119311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cognitive dysfunction is a common complication of chronic insomnia. Liuwei Anshen Capsule (LAC), a traditional Chinese patent medicine clinically prescribed for insomnia, has been proved to possess good efficacy in reducing insomnia complications including dementia and anxiety in clinic. However, the active substances in LAC and their mechanisms in treating cognitive deficit associated with sleep disorders remain unclear. AIM OF THE STUDY This study aims to explore the potential material basis and therapeutic mechanisms of LAC on cognitive impairment caused by sleep deprivation (SD) through an integrative approach involving serum pharmacochemistry, network pharmacology and experimental validation. METHODS The active ingredients of LAC in vitro and in vivo were screened and identified by liquid chromatography-mass spectrometry (LC-MS) technology. The potential targets and signaling pathways of LAC against cognitive impairment were predicted based on network pharmacology and molecular docking. Subsequently, MWM and NOR were employed to evaluate the efficacy of LAC on cognitive impairment in SD rats, and the mechanism was further validated from pathological and molecular biology perspectives. RESULTS Totally 85 active ingredients in LAC were accurately identified and 8 components absorbed into blood were found by LC-MS. Network pharmacology and molecular docking analysis predicted potential targets involving caspase-3, MAPK3, MAPK1, and Bcl-2. LAC (192, 384, and 768 mg/kg, i.g.) could improve spatial learning and memory of SD rats in a dose-dependent manner, restrain hippocampal neuronal apoptosis and microglia activation, and diminish TNF-α, IL-1β, and IL-6 expression levels, which were achieved by regulating apoptosis-related proteins (caspase-3, Bax, and Bcl-2) and MAPK (p-ERK and p-P38) signaling pathway. CONCLUSION The findings provide evidence that LAC alleviates cognitive abnormality and pathological alterations in sleep-deprived rats by regulating the expression of apoptosis related proteins and MAPK signaling pathway, indicating its potential therapy for the cognitive complaints caused by insomnia or other neurological diseases.
Collapse
Affiliation(s)
- Lian-Yu Li
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Shang-Zhi Liu
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Xuecheng Yu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiaoyuan Shi
- SCIEX, Analytical Instrument Trading Co., Ltd, Shanghai, 200355, China
| | - Hongtao You
- Chongqing Pharscin Pharmaceutical Group Co., Ltd., Chongqing, 401120, China
| | - Ping Liu
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Fei Wang
- Dept. of Brain Disease, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, 430014, China
| | - Ping Wang
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China; School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Lin-Lin Chen
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China; Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
3
|
Zhao Q, Pan Y, Zhang D, Zhou X, Sun L, Xu Z, Zhang Y. The active ingredient β-sitosterol in Ganoderma regulates CHRM2-mediated aerobic glycolysis to induce apoptosis of lung adenocarcinoma. Genes Genet Syst 2024:24-00108. [PMID: 39537174 DOI: 10.1266/ggs.24-00108] [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: 11/16/2024] Open
Abstract
BACKGROUND β-sitosterol is a natural plant steroidal compound with anti-cancer properties against various tumors. This work attempts to explore the inhibitory effect of β-sitosterol on the progression of lung adenocarcinoma (LUAD) and further analyze its targets. METHODS In this work, we applied network pharmacology to obtain the components and targets of Ganoderma spore powder. The biological functions of β-sitosterol and CHRM2 were studied using the homograft mouse model and a series of in vitro experiments including quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blot (WB), CCK-8, flow cytometry, immunohistochemistry (IHC), and immunofluorescence (IF) experiments. The regulatory influence of β-sitosterol on the glycolysis pathway was validated by detecting glucose consumption and lactate production, as well as extracellular acidification rate (ECAR) and oxygen consumption rate (OCR). RESULTS In this project, we unearthed that CHRM2 was a protein that directly binds to β-sitosterol. In vitro, CHRM2 overexpression repressed the apoptosis rate and expression of apoptosis-related proteins and promoted glycolysis, while the addition of lonidamine attenuated the inhibitory effect conferred by CHRM2 overexpression on LUAD apoptosis. Furthermore, β-sitosterol hindered glycolysis as well as the growth of tumors in vitro and in vivo. CHRM2 overexpression reversed the effect of β-sitosterol on the biological behavior of LUAD cells. CONCLUSION Our project emphasized that CHRM2 is a direct target of β-sitosterol in LUAD cells. β-sitosterol can repress the glycolysis pathway, exerting an anti-tumor effect. These findings can provide new evidence for supporting the potential use of β-sitosterol as a therapeutic agent for LUAD.
Collapse
Affiliation(s)
- Qiong Zhao
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College
| | - Yuting Pan
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College
| | - Danjia Zhang
- Department of Traditional Chinese Medicine, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College
| | - Xiaolian Zhou
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College
| | - Liangyun Sun
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College
| | - Zihan Xu
- MPA, Cornell University, Brooks School
| | | |
Collapse
|
4
|
Liu M, Yang K, Qiu H. Exploring the Effect of Gomisin A on Non-Small Cell Lung Cancer With Network Pharmacology, Molecular Docking, In Vitro and In Vivo Assays. Chem Biol Drug Des 2024; 104:e70014. [PMID: 39533773 DOI: 10.1111/cbdd.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/08/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Gomisin A is an active ingredient of Schisandra chinensis. Pre-clinical studies suggest Gomisin A has good anti-cancer activities against a variety of cancers, but its mechanism of action in non-small cell lung cancer (NSCLC) is unclear. This study aims to explore the potential mechanism of Gomisin A in treating NSCLC. The SwissTargetPrediction, CTD, HERB and PharmMapper databases were used to collect related targets of Gomisin A. NSCLC-related genes were obtained using the GEO, CTD, DisGeNET, OMIM, GeneCards, NCBI, and PharmGKB databases. The central targets and potential mechanisms of Gomisin A against NSCLC were screened using network pharmacology and molecular docking. Finally, the therapeutic activity of Gomisin A on NSCLC was verified by experiments. A total of 161 potential targets of Gomisin A against NSCLC were identified. TNF, AKT1, STAT3, and IL6 were identified as the central targets of Gomisin A. The binding energy of Gomisin A and the central targets was less than -5 kcal/mol. Gomisin A could inhibit NSCLC cell viability, migration and invasion and induce cell cycle arrest and apoptosis. Gomisin A also inhibited in vivo metastasis of NSCLC cells. In addition, Gomisin A could also reduce the expression level of the central targets and inhibit the PI3K-Akt signaling pathway. In summary, Gomisin A may be a candidate drug for the treatment of NSCLC, and TNF, AKT1, STAT3, and IL6 are potential targets for Gomisin A in NSCLC treatment, and its therapeutic mechanism may be related to the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Mei Liu
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Kai Yang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Huibing Qiu
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
5
|
Zhang F, Gu T, Li J, Zhu Y, Chu M, Zhou Q, Liu J. Emodin regulated lactate metabolism by inhibiting MCT1 to delay non-small cell lung cancer progression. Hum Cell 2024; 38:11. [PMID: 39465441 DOI: 10.1007/s13577-024-01140-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024]
Abstract
Lung cancer is one of the most common malignant tumors in the world, with high incidence rate and mortality. Monocarboxylate transporter (MCT) 1 has been found to be widely expressed in various tumors and plays a crucial role in regulating energy metabolism. Emodin, as an important traditional Chinese medicine in China, has been reported to inhibit the progression of lung cancer. However, its potential mechanism has not been fully elucidated. The effects of emodin and MCT1 inhibitor AZD3965 on the proliferation, migration, and invasion of lung cancer cells were detected using cell counting kit-8 (CCK-8) assay, wound-healing assay, and transwell small chamber assay. The content of glucose, lactate, and pyruvate in the cell culture medium was detected using a glucose, lactate, and pyruvate detection kit, and also detected protein expression using western blotting. In addition, to investigate the effects of emodin and AZD3965 on lung cancer in vivo, we constructed nude mice subcutaneous transplant tumor model by subcutaneous injection of lung cancer cells. The results showed that emodin and AZD3965 could inhibit the proliferation, migration, and invasion of lung cancer cells. At the same time, they could inhibit the expression of MCT1 in lung cancer cells and promote the release of lactate, but did not affect the content of glucose and pyruvate. In vivo experiments had shown that emodin and AZD3965 could effectively inhibit the growth of lung cancer and inhibit the expression of MCT1. All in all, our data suggested that emodin inhibited the proliferation, migration, and invasion of lung cancer cells, possibly by inhibiting MCT1, providing important theoretical basis for elucidating the mechanism of emodin in treating lung cancer.
Collapse
Affiliation(s)
- Fei Zhang
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Tian Gu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Jin Li
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Yanqiu Zhu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Mingliang Chu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Qing Zhou
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Jiemin Liu
- Department of Endoscopy, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
6
|
Wang L, Chen H, Deng L, Hu M, Wang Z, Zhang K, Lian C, Wang X, Zhang J. Roburic acid inhibits lung cancer metastasis and triggers autophagy as verified by network pharmacology, molecular docking techniques and experiments. Front Oncol 2024; 14:1449143. [PMID: 39450260 PMCID: PMC11499198 DOI: 10.3389/fonc.2024.1449143] [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: 06/22/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Background Roburic acid (ROB) is a newly discovered tetracyclic triterpene acid extracted from oak galls, which has anti-inflammatory effects, but the mechanism of its anticancer effect is not clear. Our study focuses on exploring the potential mechanism of action of ROB in the treatment of lung cancer using a combination of network pharmacological prediction, molecular docking technique and experimental validation. Methods A network pharmacology approach was used to screen the protein targets of ROB and lung cancer, and PPI network analysis and enrichment analysis were performed on the intersecting genes. The tissue and organ distribution of the targets was also evaluated based on the BioGPS database. To ensure the reliability of the network pharmacology prediction results, we proceeded to use molecular docking technique to determine the relationship between drugs and targets. Finally, in vitro experiments with cell lines were performed to further reveal the potential mechanism of ROB for the treatment of lung cancer. Results A total of 83 potential targets of ROB in lung cancer were collected and further screened by using Cytoscape software, and 7 targets of PTGS2, CYP19A1, PTGS1, AR, CYP17A1, PTGES and SRD5A1 were obtained as hub genes and 7 hub targets had good binding energy with ROB. GO and KEGG analysis showed that ROB treatment of lung cancer mainly involves Arachidonic acid metabolism, Notch signaling pathway, cancer pathway and PPAR signaling pathway. The results of in vitro experiments indicated that ROB may inhibit the proliferation and metastasis of lung cancer cells and activate the PPARγ signaling pathway, as well as induce cellular autophagy. Conclusions The results of this study comprehensively elucidated the potential targets and molecular mechanisms of ROB for the treatment of lung cancer, providing new ideas for further lung cancer therapy.
Collapse
Affiliation(s)
- Luyao Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Lili Deng
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Mengling Hu
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Kai Zhang
- Research Center of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Chaoqun Lian
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Joint Research Center for Regional Diseases of Institute of Healthcare Management (IHM), The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| |
Collapse
|
7
|
De L, Xing N, Du Q, Guo S, Wang S. Investigating the anti-lung cancer properties of Zhuang medicine Cycas revoluta Thunb. leaves targeting ion channels and transporters through a comprehensive strategy. Comput Biol Chem 2024; 112:108156. [PMID: 39067352 DOI: 10.1016/j.compbiolchem.2024.108156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/24/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Cycas revoluta Thunb., known for its ornamental, economic, and medicinal value, has leaves often discarded as waste. However, in ethnic regions of China, the leaves (CRL) are used in folk medicine for anti-tumor properties, particularly for regulating pathways related to cancer. Recent studies on ion channels and transporters (ICTs) highlight their therapeutic potential against cancer, making it vital to identify CRL's active constituents targeting ICTs in lung cancer. PURPOSE This study aims to uncover bioactive substances in CRL and their mechanisms in regulating ICTs for lung cancer treatment using network pharmacology, bioinformatics, molecular docking, molecular dynamics (MD) simulations, in vitro cell assays and HPLC. METHODS We analyzed 62 CRL compounds, predicted targets using PubChem and SwissTargetPrediction, identified lung cancer and ICT targets via GeneCards, and visualized overlaps with R software. Interaction networks were constructed using Cytoscape and STRING. Gene expression, GO, and KEGG analyses were performed using R software. TCGA data provided insights into differential, correlation, survival, and immune analyses. Key interactions were validated through molecular docking and MD simulations. Main biflavonoids were quantified using HPLC, and in vitro cell viability assays were conducted for key biflavonoids. RESULTS Venn diagram analysis identified 52 intersecting targets and ten active CRL compounds. The PPI network highlighted seven key targets. GO and KEGG analysis showed CRL-targeted ICTs involved in synaptic transmission, GABAergic synapse, and proteoglycans in cancer. Differential expression and correlation analysis revealed significant differences in five core targets in lung cancer tissues. Survival analysis linked EGFR and GABRG2 with overall survival, and immune infiltration analysis associated the core targets with most immune cell types. Molecular docking indicated strong binding of CRL ingredients to core targets. HPLC revealed amentoflavone as the most abundant biflavonoid, followed by hinokiflavone, sciadopitysin, and podocarpusflavone A. MD simulations showed that podocarpusflavone A and amentoflavone had better binding stability with GABRG2, and the cell viability assay also proved that they had better anti-lung cancer potential. CONCLUSIONS This study identified potential active components, targets, and pathways of CRL-targeted ICTs for lung cancer treatment, suggesting CRL's utility in drug development and its potential beyond industrial waste.
Collapse
Affiliation(s)
- Luo De
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Meishan Traditional Chinese Medicine Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qinyun Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Sa Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Meishan Traditional Chinese Medicine Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
8
|
Yang C, Zhao M, Chen Y, Song J, Wang D, Zou M, Liu J, Wen W, Xu S. Dietary bitter ginger-derived zerumbone improved memory performance during aging through inhibition of the PERK/CHOP-dependent endoplasmic reticulum stress pathway. Food Funct 2024; 15:9070-9084. [PMID: 39078275 DOI: 10.1039/d4fo00402g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
PERK/CHOP pathway-mediated excessive endoplasmic reticulum (ER) stress is closely linked to aging-related cognitive impairment (ARCD). Zerumbone (ZB), a naturally occurring sesquiterpene molecule obtained from dietary bitter ginger, has garnered significant interest due to its diverse range of biological properties. It is unclear, though, if ZB can reduce ARCD by preventing ER stress that is dependent on the PERK/CHOP pathway. Here, the PERK-CHOP ER stress pathway was the main focus of an evaluation of the effects and mechanisms of ZB for attenuating ARCD in D-galactose (D-gal)-induced aging mice and SH-SY5Y cells. According to our findings, ZB not only greatly decreased neuronal impairment both in vitro and in vivo, but also significantly alleviated learning and memory failure in vivo. ZB significantly reduced the activation of the PERK/CHOP pathway and neuronal apoptosis in vitro and in vivo, exhibiting the down-regulation of GRP78, p-PREK/PERK, and CHOP expression levels, in addition to suppressing oxidative damage (MDA drop and SOD rise). Comparable outcomes were noted in SH-SY5Y cells subjected to severe ER stress caused by TM. On the other hand, 4-PBA, an ER stress inhibitor, considerably reversed these modifications. Remarkably, CCT020312 (a PERK activator) dramatically overrode the inhibitory effects of ZB on the PERK/CHOP pathway and neuronal death in D-gal-induced SH-SY5Y cells. In contrast, GSK2606414 (a PERK inhibitor) significantly increased these effects of ZB. In summary, our results suggested that ZB prevented D-gal-induced cognitive deficits by blocking the PERK/CHOP-dependent ER stress pathway and apoptosis, suggesting that ZB might be a natural sesquiterpene molecule that relieves ARCD.
Collapse
Affiliation(s)
- Chuan Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Meihuan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuanyuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Juxian Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dan Wang
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Mi Zou
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jingru Liu
- University College London, Gower Street, London WC1E 6BT, UK
| | - Wen Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Institute of Materia Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
9
|
Zhang W, Cai S, Qin L, Feng Y, Ding M, Luo Z, Shan J, Di L. Alkaloids of Aconiti Lateralis Radix Praeparata inhibit growth of non-small cell lung cancer by regulating PI3K/Akt-mTOR signaling and glycolysis. Commun Biol 2024; 7:1118. [PMID: 39261597 PMCID: PMC11390937 DOI: 10.1038/s42003-024-06801-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Aconiti Lateralis Radix Praeparata (Fuzi in Chinese) is widely used in the clinical treatment of tumors. This study aims to explore the active fractions and underlying mechanisms of Fuzi in the treatment of non-small cell lung cancer (NSCLC). Fuzi alkaloids (FZA) is prepared and found to inhibit the growth of NSCLC both in vitro and in vivo significantly. A total of 53 alkaloids are identified in FZA by UPLC-Q-TOF-MS. Proteomics experiment show that 238 differentially expressed proteins regulated by FZA are involved in amino acid anabolism, pyrimidine metabolism and PI3K/Akt-mTOR signaling pathway. Metabolomics analyses identify 32 significant differential metabolites which are mainly involved in amino acid metabolism, TCA cycle and other pathways. Multi-omics research combined with molecular biological assays suggest that FZA might regulate glycolysis through PI3K/Akt-mTOR pathway to treat NSCLC. The study lays a foundation for the anti-cancer investigation of Fuzi and provides a possible scientific basis for its clinical application.
Collapse
Affiliation(s)
- Wen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| | - Shuhui Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Lihong Qin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Yaru Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Menglei Ding
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Zichen Luo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| |
Collapse
|
10
|
Yu J, Song H, Zhou L, Wang S, Liu X, Liu L, Ma Y, Li L, Wen S, Luo Y, Zhang X, Li W, Niu X. (-)-Epicatechin gallate prevented atherosclerosis by reducing abnormal proliferation of VSMCs and oxidative stress of AML 12 cells. Cell Signal 2024; 121:111276. [PMID: 38936786 DOI: 10.1016/j.cellsig.2024.111276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
(-)-Epicatechin gallate (ECG) is beneficial to the treatment of cardiovascular diseases (CVDs), especially atherosclerosis (AS) through antioxidant stress, but there is a lack of detailed mechanism research. In this study, the therapeutic target of ECG was determined by crossing the drug target and disease target of CVDs and AS. The combination ability of ECG with important targets was verified by Discovery Studio software. The abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by Ang-II and the oxidative damage of AML 12 induced by H2O2 were established to verify the reliability of ECG intervention on the target protein. A total of 120 ECG targets for the treatment of CVDs-AS were predicted by network pharmacology. The results of molecular docking showed that ECG has strong binding force with VEGFA, MMP-9, CASP3 and MMP-2 domains. In vitro experiments confirmed that ECG significantly reduced the expression of VEGFA, MMP-9, CASP3 and MMP-2 in Ang-II-induced VSMCs, and also blocked the abnormal proliferation, oxidative stress and inflammatory reaction of VSMCs by inhibiting the phosphorylation of PI3K signaling pathway. At the same time, ECG also interfered with H2O2-induced oxidative damage of AML 12 cells, decreased the expression of ROS and MDA and cell foaming, and increased the activities of antioxidant enzymes such as SOD, thus playing a protective role.
Collapse
Affiliation(s)
- Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lingyi Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajing Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lingli Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Sha Wen
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yuzhi Luo
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinya Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
11
|
Li Y, Yin Y, Xiong J, Zhang Z, Li L, Zhang B, Zhang F, Mao D. Combining Network Pharmacology and Transcriptomics to Investigate the Mechanisms of Yujiang Paidu Decoction in the Treatment of Chronic Rhinosinusitis with Nasal Polyps. Drug Des Devel Ther 2024; 18:3791-3809. [PMID: 39219695 PMCID: PMC11365509 DOI: 10.2147/dddt.s461769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Background Yujiang Paidu Decoction (YJPD) has demonstrated clinical efficacy in the treatment of chronic rhinosinusitis. However, the effects and mechanisms of the YJPD on chronic rhinosinusitis with nasal polyps (CRSwNP) remain unclear. Purpose This study aimed to elucidate the potential mechanism of action of YJPD in the treatment of CRSwNP based on network pharmacology, transcriptomics and experiments. Methods A CRSwNP mouse model was established using ovalbumin (OVA) and staphylococcus aureus enterotoxin B (SEB) for 12 weeks and the human nasal epithelial cell (HNEpC) model was induced with IL-13 in vitro. Behavioral tests, scanning electron microscopy (SEM), micro-CT and pathological change of nasal tissues were observed to investigate the therapeutic effects of YJPD. Network pharmacology and transcriptomics were launched to explore the pharmacological mechanisms of YJPD in CRSwNP treatment. Finally, an ELISA, immunofluorescence, RT-qPCR, Western blotting and Tunel were performed for validation. Results Different doses of YJPD intervention effectively alleviated rubbing and sneezing symptoms in CRSwNP mice. Additionally, YJPD significantly reduced abnormal serological markers, structural damage of the nasal mucosa, inflammatory cell infiltration, goblet cell increases, and inhibited OVA-specific IgE levels and the secretion of Th2 cytokines such as IL-4, IL-5, and IL-13. Moreover, transcriptomics and network pharmacology analyses indicated that YJPD may exert anti-inflammatory and anti-apoptotic effects by inhibiting the MAPK/AP-1 signaling pathway. The experimental findings supported this conclusion, which was further corroborated by similar results observed in IL13-induced HNEpCs in vitro. Conclusion YJPD could alleviate inflammatory status and epithelial apoptosis by inhibiting aberrant activation of MAPK/AP-1 signaling pathway. This finding provides a strong basis for using YJPD as a potential treatment in CRSwNP.
Collapse
Affiliation(s)
- Yujie Li
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yadong Yin
- Xijing Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Juan Xiong
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhipeng Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China
| | - Linglong Li
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, People’s Republic of China
| | - Baoshun Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China
| | - Feng Zhang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, People’s Republic of China
| | - Dehong Mao
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
12
|
Wang R, Gao C, Yu M, Song J, Feng Z, Wang R, Pan H, Liu H, Li W, Fan X. Mechanistic prediction and validation of Brevilin A Therapeutic effects in Lung Cancer. BMC Complement Med Ther 2024; 24:214. [PMID: 38840248 PMCID: PMC11151568 DOI: 10.1186/s12906-024-04516-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Traditional Chinese medicine (TCM) has been found widespread application in neoplasm treatment, yielding promising therapeutic candidates. Previous studies have revealed the anti-cancer properties of Brevilin A, a naturally occurring sesquiterpene lactone derived from Centipeda minima (L.) A.Br. (C. minima), a TCM herb, specifically against lung cancer. However, the underlying mechanisms of its effects remain elusive. This study employs network pharmacology and experimental analyses to unravel the molecular mechanisms of Brevilin A in lung cancer. METHODS The Batman-TCM, Swiss Target Prediction, Pharmmapper, SuperPred, and BindingDB databases were screened to identify Brevilin A targets. Lung cancer-related targets were sourced from GEO, Genecards, OMIM, TTD, and Drugbank databases. Utilizing Cytoscape software, a protein-protein interaction (PPI) network was established. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene set enrichment analysis (GSEA), and gene-pathway correlation analysis were conducted using R software. To validate network pharmacology results, molecular docking, molecular dynamics simulations, and in vitro experiments were performed. RESULTS We identified 599 Brevilin A-associated targets and 3864 lung cancer-related targets, with 155 overlapping genes considered as candidate targets for Brevilin A against lung cancer. The PPI network highlighted STAT3, TNF, HIF1A, PTEN, ESR1, and MTOR as potential therapeutic targets. GO and KEGG analyses revealed 2893 enriched GO terms and 157 enriched KEGG pathways, including the PI3K-Akt signaling pathway, FoxO signaling pathway, and HIF-1 signaling pathway. GSEA demonstrated a close association between hub genes and lung cancer. Gene-pathway correlation analysis indicated significant associations between hub genes and the cellular response to hypoxia pathway. Molecular docking and dynamics simulations confirmed Brevilin A's interaction with PTEN and HIF1A, respectively. In vitro experiments demonstrated Brevilin A-induced dose- and time-dependent cell death in A549 cells. Notably, Brevilin A treatment significantly reduced HIF-1α mRNA expression while increasing PTEN mRNA levels. CONCLUSIONS This study demonstrates that Brevilin A exerts anti-cancer effects in treating lung cancer through a multi-target and multi-pathway manner, with the HIF pathway potentially being involved. These results lay a theoretical foundation for the prospective clinical application of Brevilin A.
Collapse
Affiliation(s)
- Ruixue Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Cuiyun Gao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Meng Yu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jialing Song
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Zhenzhen Feng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ruyu Wang
- School of clinical medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haimeng Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Wei Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xiangzhen Fan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
13
|
Tan B, Lan X, Zhang Y, Liu P, Jin Q, Wang Z, Liang Z, Song W, Xuan Y, Sun Y, Li Y. Effect of 23‑hydroxybetulinic acid on lung adenocarcinoma and its mechanism of action. Exp Ther Med 2024; 27:239. [PMID: 38633355 PMCID: PMC11019653 DOI: 10.3892/etm.2024.12527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/12/2024] [Indexed: 04/19/2024] Open
Abstract
The present study aimed to investigate the effect and mechanism of Pulsatilla compounds on lung adenocarcinoma. The representative drug chosen was the compound 23-HBA. GeneCards, Swiss target prediction, DisGeNET and TCMSP were used to screen out related genes, and MTT and flow cytometry assays were used to verify the inhibitory effect of Pulsatilla compounds on the proliferation of lung adenocarcinoma cells. Subsequently, the optimal target, peroxisome proliferator-activated receptor (PPAR)-γ, was selected using bioinformatics analysis, and its properties of low expression in lung adenocarcinoma cells and its role as a tumor suppressor gene were verified by western blot assay. The pathways related to immunity and inflammation, vascular function, cell proliferation, differentiation, development and apoptosis with the highest degree of enrichment and the mechanisms were explored through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. Finally, the clinical prognosis in terms of the survival rate of patients in whom the drug is acting on the target was analyzed using the GEPIA database. The results indicated that Pulsatilla compounds can inhibit the proliferation of lung adenocarcinoma cells by blocking the cell cycle at the G1 phase. Subsequently, the related PPAR-γ gene was verified as a tumor suppressor gene. Further analysis demonstrated that this finding was related to the PPAR signaling pathway and mitochondrial reactive oxygen species (ROS) production. Finally, the clinical prognosis was found to be improved, as the survival rate of patients was increased. In conclusion, Pulsatilla compounds were indicated to inhibit the viability and proliferation of lung adenocarcinoma H1299 cells, and the mechanism of action was related to PPAR-γ, the PPAR signaling pathway and mitochondrial ROS. The present study provides novel insight to further explore the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Boyu Tan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaoxu Lan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yifan Zhang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pai Liu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Qiyao Jin
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhiqiang Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhidong Liang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wei Song
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ye Xuan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yunxiao Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
14
|
Yuan J, Lin M, Yang S, Yin H, Ouyang S, Xie H, Tang H, Ou X, Zeng Z. The therapeutic effect and targets of herba Sarcandrae on breast cancer and the construction of a prognostic signature consisting of inflammation-related genes. Heliyon 2024; 10:e31137. [PMID: 38778969 PMCID: PMC11109893 DOI: 10.1016/j.heliyon.2024.e31137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Background The prevalence of breast cancer (BRCA), which is common among women, is on the rise. This study applied network pharmacology to explore the potential mechanism of action of herba sarcandrae in BRCA and construct a prognostic signature composed of inflammation-related genes. Methods The active ingredients of herba sarcandrae were screened using the SymMap, TCMID, and TCMSP platforms, and the molecular targets were determined in the UniProt database. The "drug-active compound-potential target" network was established with Cytoscape 3.7.2. The molecular targets were subjected to disease ontology, gene ontology (GO), and Kyoto Encyclopedia of Genes (KEGG) analyses. AutoDock software was used for molecular docking. Differentially expressed genes (DEGs) related to inflammation were obtained from the BRCA Cancer Genome Atlas (TCGA) database. In the training cohort, the univariate Cox regression model was applied to preliminarily screen prognostic genes. A multigene signature was built by the least absolute shrinkage and selection operator (LASSO) regression model, followed by validation through Kaplan‒Meier, Cox, and receiver operating characteristic (ROC) analyses. Results Forty-one active compounds were identified, and 265 therapeutic targets for herba sarcandrae were predicted. GO enrichment results revealed significant enrichment of biological processes, such as response to xenobiotic stimuli, response to nutrient levels, and response to lipopolysaccharide. KEGG analysis revealed significant enrichment of pathways such as AGE-RAGE and chemical carcinogenesis receptor activation signaling pathways. In addition, the herbs Marc-Andre and rutin were shown to mediate BRCA cell proliferation and apoptosis via the interferon regulatory factor 1 (IRF1)/signal transducer and activator of transcription 3 (STAT3)/programmed death-ligand 1 (PD-L1) pathway. Sixteen inflammatory signatures, including BST2, GPR132, IL12B, IL18, IL1R1, IL2RB, IRF1, and others, were constructed, and the risk score was found to be a strong independent prognostic factor for overall survival in BRCA patients. The 16-inflammation signature was associated with several clinical features (age, clinical stage, T, and N classifications) and could reflect immune cell infiltration in tumor microenvironments with different immune cells. Conclusions Herba sarcandrae and rutin were shown to mediate BRCA cell proliferation and apoptosis via the IRF1/STAT3/PD-L1 pathway, and the 16-member inflammatory signature might be a novel biomarker for predicting BRCA patient prognosis, providing more accurate guidance for clinical treatment prognosis evaluation and having important reference value for individualized treatment selection.
Collapse
Affiliation(s)
- Jie Yuan
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Minxia Lin
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Shaohua Yang
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hao Yin
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Shaoyong Ouyang
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hong Xie
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hongmei Tang
- Pharmaceutical Department, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaowei Ou
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Zhiqiang Zeng
- Department of General Surgery, Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| |
Collapse
|
15
|
Zhang W, Ding M, Feng Y, Cai S, Luo Z, Shan J, Di L. Modulation of cellular metabolism and alleviation of bacterial dysbiosis by Aconiti Lateralis Radix Praeparata in non-small cell lung cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155099. [PMID: 38412665 DOI: 10.1016/j.phymed.2023.155099] [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: 06/25/2023] [Revised: 08/22/2023] [Accepted: 09/17/2023] [Indexed: 02/29/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a highly prevalent and fatal form of lung cancer. In China, Aconiti Lateralis Radix Praeparata (Fuzi in Chinese), derived from the lateral root of Aconitum carmichaeli Debx. (Ranunculaceae, Aconitum), is extensively prescribed to treat cancer in traditional medicine and clinical practice. However, the precise mechanism by which Fuzi treats NSCLC remains unknown. PURPOSE This article aims to assess the efficacy of Fuzi against NSCLC and elucidate its underlying mechanism. METHODS Marker ingredients of Fuzi decoction were quantified using UPLC-TSQ-MS. The effectiveness of Fuzi on NSCLC was evaluated using a xenograft mouse model. Subsequently, a comprehensive approach involving network pharmacology, serum metabolomics, and 16S rDNA sequencing was employed to investigate the anti-NSCLC mechanism of Fuzi. RESULTS Pharmacological evaluation revealed significant tumour growth inhibition by Fuzi, accompanied by minimal toxicity. Network pharmacology identified 29 active Fuzi compounds influencing HIF-1, PI3K/Akt signalling, and central carbon metabolism in NSCLC. Integrating untargeted serum metabolomics highlighted 30 differential metabolites enriched in aminoacyl-tRNA biosynthesis, alanine, aspartate, and glutamate metabolism, and the tricarboxylic acid (TCA) cycle. Targeted serum metabolomics confirmed elevated glucose content and reduced levels of pyruvate, lactate, citrate, α-ketoglutarate, succinate, fumarate, and malate following Fuzi administration. Furthermore, 16S rDNA sequencing assay showed that Fuzi ameliorated the dysbiosis after tumorigenesis, decreased the abundance of Proteobacteria, and increased that of Firmicutes and Bacteriodetes. PICRUSt analysis revealed that Fuzi modulated the pentose phosphate pathway of the gut microbiota. Spearman correlation showed that Proteobacteria and Escherichia_Shigella accelerated the TCA cycle, whereas Bacteroidota, Bacteroides, and Lachnospiraceae_NK4A136_group suppressed the TCA cycle. CONCLUSIONS This study firstly introduces a novel NSCLC mechanism involving Fuzi, encompassing energy metabolism and intestinal flora. It clarifies the pivotal role of the gut microbiota in treating NSCLC and modulating the TCA cycle. Moreover, these findings offer valuable insights for clinical practices and future research of Fuzi against NSCLC.
Collapse
Affiliation(s)
- Wen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| | - Menglei Ding
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China; Department of Pharmacy, Kunshan Hospital of Chinese Medicine, Suzhou, China
| | - Yaru Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Shuhui Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Zichen Luo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| |
Collapse
|
16
|
Huang P, Wang Y, Liu C, Zhang Q, Ma Y, Liu H, Wang X, Wang Y, Wei M, Ma L. Exploring the Mechanism of Zhishi-Xiebai-Guizhi Decoction for the Treatment of Hypoxic Pulmonary Hypertension based on Network Pharmacology and Experimental Analyses. Curr Pharm Des 2024; 30:2059-2074. [PMID: 38867532 DOI: 10.2174/0113816128293601240523063527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/26/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Hypoxic Pulmonary Hypertension (HPH), a prevalent disease in highland areas, is a crucial factor in various complex highland diseases with high mortality rates. Zhishi-Xiebai-Guizhi decoction (ZXGD), traditional Chinese medicine with a long history of use in treating heart and lung diseases, lacks a clear understanding of its pharmacological mechanism. OBJECTIVE This study aimed to investigate the pharmacological effects and mechanisms of ZXGD on HPH. METHODS We conducted a network pharmacological prediction analysis and molecular docking to predict the effects, which were verified through in vivo experiments. RESULTS Network pharmacological analysis revealed 51 active compounds of ZXGD and 701 corresponding target genes. Additionally, there are 2,116 targets for HPH, 311 drug-disease co-targets, and 17 core-targets. GO functional annotation analysis revealed that the core targets primarily participate in biological processes such as apoptosis and cellular response to hypoxia. Furthermore, KEGG pathway enrichment analysis demonstrated that the core targets are involved in several pathways, including the phosphatidylinositol-3 kinase/protein kinase B (PI3K/Akt) signaling pathway and Hypoxia Inducible Factor 1 (HIF1) signaling pathway. In vivo experiments, the continuous administration of ZXGD demonstrated a significant improvement in pulmonary artery pressure, right heart function, pulmonary vascular remodeling, and pulmonary vascular fibrosis in HPH rats. Furthermore, ZXGD was found to inhibit the expression of PI3K, Akt, and HIF1α proteins in rat lung tissue. CONCLUSION In summary, this study confirmed the beneficial effects and mechanism of ZXGD on HPH through a combination of network pharmacology and in vivo experiments. These findings provided a new insight for further research on HPH in the field of traditional Chinese medicine.
Collapse
Affiliation(s)
- Pan Huang
- Qinghai University Medical College, Xining 810016, China
| | - Yuxiang Wang
- Qinghai University Medical College, Xining 810016, China
| | - Chuanchuan Liu
- Hydatidosis Laboratory, Affiliated Hospital of Qinghai University, Xining 810012, China
| | - Qingqing Zhang
- Qinghai University Medical College, Xining 810016, China
| | - Yougang Ma
- Qinghai University Medical College, Xining 810016, China
| | - Hong Liu
- Qinghai University Medical College, Xining 810016, China
| | - Xiaobo Wang
- Qinghai University Medical College, Xining 810016, China
| | - Yating Wang
- Qinghai University Medical College, Xining 810016, China
| | - Minmin Wei
- Qinghai University Medical College, Xining 810016, China
- Qinghai Provincial Hospital of Traditional Chinese Medicine, Xining 810099, China
| | - Lan Ma
- Qinghai University Medical College, Xining 810016, China
| |
Collapse
|
17
|
Chen R, Song C, Qiu J, Su Q, Wang X, Deng G, Cheng K, Chen X, Xiang W, Liu T, Chen X, Wu J. Exploring the potential mechanism of Taohong Siwu decoction in the treatment of avascular necrosis of the femoral head based on network pharmacology and molecular docking. Medicine (Baltimore) 2023; 102:e35312. [PMID: 38115279 PMCID: PMC10727545 DOI: 10.1097/md.0000000000035312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 12/21/2023] Open
Abstract
Based on network pharmacology and molecular docking, this study seeks to investigate the mechanism of Taohong Siwu decoction (THSWD) in the treatment of avascular necrosis of the femoral head (AVNFH). The Traditional Chinese Medicine Systems Pharmacology database was used in this investigation to obtain the active ingredients and related targets for each pharmaceutical constituent in THSWD. To find disease-related targets, the terms "avascular necrosis of the femoral head," "necrosis of the femoral head," "steroid-induced necrosis of the femoral head," "osteonecrosis," and "avascular necrosis of the bone" were searched in the databases DisGeNET, GeneCards, Comparative Toxicogenomics Database, and MalaCards. Following the identification of the overlap targets of THSWD and AVNFH, enrichment analysis using gene ontology, Kyoto Encyclopedia of Genes and Genomes, Reactome, and WikiPathways was conducted. The "THSWD-drug-active compound-intersection gene-hub gene-AVNFH" network and protein-protein interaction network were built using Cytoscape 3.9.1 and string, and CytoHubba was used to screen hub genes. The binding activities of hub gene targets and key components were confirmed by molecular docking. 152 prospective therapeutic gene targets were found in the bioinformatics study of ONFH treated with THSWD, including 38 major gene targets and 10 hub gene targets. The enrichment analysis of 38 key therapeutic targets showed that the biological process of gene ontology analysis mainly involved cytokine-mediated signaling pathway, angiogenesis, cellular response to reactive oxygen species, death-inducing signaling complex. The Kyoto Encyclopedia of Genes and Genomes signaling pathway mainly involves TNF signaling pathway, IL-17 signaling pathway, and the Recactome pathway mainly involves Signaling by Interleukins, Apoptosis, and Intrinsic Pathway for Apoptosis. WikiPathways signaling pathway mainly involves TNF-related weak inducer of apoptosis signaling pathway, IL-18 signaling pathway. According to the findings of enrichment analysis, THSWD cured AVNFH by regulating angiogenesis, cellular hypoxia, inflammation, senescence, apoptosis, cytokines, and cellular proliferation through the aforementioned targets and signaling pathways. The primary component of THSWD exhibits a strong binding force with the key protein of AVNFH. This study sheds new light on the biological mechanism of THSWD in treating AVNFH by revealing the multi-component, multi-target, and multi-pathway features and molecular docking mechanism of THSWD.
Collapse
Affiliation(s)
- Rui Chen
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chao Song
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Junjie Qiu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qifan Su
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaoqiang Wang
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Guanghui Deng
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Kang Cheng
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaoyu Chen
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Wei Xiang
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Tao Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaojun Chen
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiaqi Wu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
18
|
Li H, Shi W, Shen T, Hui S, Hou M, Wei Z, Qin S, Bai Z, Cao J. Network pharmacology-based strategy for predicting therapy targets of Ecliptae Herba on breast cancer. Medicine (Baltimore) 2023; 102:e35384. [PMID: 37832105 PMCID: PMC10578738 DOI: 10.1097/md.0000000000035384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/04/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer is a prevalent malignancy affecting women globally, characterized by significant morbidity and mortality rates. Ecliptae Herba is a traditional herbal medicine commonly used in clinical practice, has recently been found to possess antitumor properties. In order to explore the underlying material basis and molecular mechanisms responsible for the anti-breast cancer effects of Ecliptae Herba, we used network pharmacology and experimental verification. UPLC-MS/MS was utilized to identify compounds present in Ecliptae Herba. The active components of Ecliptae Herba and its breast cancer targets were screened using public databases. Hub genes were identified using the STRING and Metascape database. The R software was utilized for visual analysis of GO and KEGG pathways. The affinity of the hub targets for the active ingredients was assessed by molecular docking analysis, which was verified by experimental assessment. A total of 178 targets were obtained from the 10 active components of Ecliptae Herba, while 3431 targets associated with breast cancer were screened. There were 144 intersecting targets between the components and the disease. Targets with a higher degree, namely EGFR and TGFB1, were identified through the hub subnetwork of PPI. GO and KEGG analyses revealed that Ecliptae Herba plays an important role in multiple cancer therapeutic mechanisms. Moreover, molecular docking results showed that the core components had good binding affinity with key targets. Finally, it was confirmed that TGF-β1 might be a potential crucial target of Ecliptae Herba in the treatment of breast cancer by cytological experiments, and the TGF-β1/Smad signaling pathway might be an important pathway for Ecliptae Herba to exert its therapeutic effects. This study elucidated the active ingredients, key targets, and molecular mechanisms of Ecliptae Herba in the treatment of breast cancer, providing a scientific foundation and therapeutic mechanism for the prevention and treatment of breast cancer with Traditional Chinese medicine.
Collapse
Affiliation(s)
- Hui Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wei Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tingming Shen
- Ningde Hospital of Traditional Chinese Medicine, Ningde, China
| | - Siwen Hui
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Manting Hou
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ziying Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junling Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Luoyang Branch of Dongzhimen Hospital Afiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|