1
|
Qiu J, Xiao G, Yang M, Huang X, Cai D, Xie C, Chen Z, Bi X, Xu A. Integrated network pharmacology and metabolomics reveal the mechanisms of Jasminum elongatum in anti-ulcerative colitis. Sci Rep 2023; 13:22449. [PMID: 38105335 PMCID: PMC10725889 DOI: 10.1038/s41598-023-49792-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
Jasminum elongatum (JE), an ethnic Chinese medicine, is widely used in the Lingnan region of China, because of its analgesic and antidiarrheal action, as well as its anti-inflammatory effects in gastrointestinal diseases. However, whether JE could against ulcerative colitis (UC) remains unclear. This research aims to reveal JE in treating UC and clarify the underlying mechanism. We used the 2.5% dextran sulfate sodium (DSS)-induced UC mice (C57BL/6J) to evaluate the therapeutic effects of JE. Metabolomics of serum and network pharmacology were combined to draw target-metabolite pathways. Apart from that, the targets of associated pathways were confirmed, and the mechanism of action was made clear, using immunohistochemistry. The pharmacodynamic results, including disease activity index (DAI), histological evaluation, and inflammatory cytokines in colon tissues, demonstrated that JE significantly relieved the physiological and pathological symptoms of UC. Network pharmacology analysis indicated 25 core targets, such as TNF, IL-6, PTGS2 and RELA, and four key pathways, including the NF-κB signaling pathway and arachidonic acid metabolism pathway, which were the key connections between JE and UC. Metabolomics analysis identified 45 endogenous differential metabolites and 9 metabolic pathways by enrichment, with the arachidonic acid metabolism pathway being the main metabolism pathway, consistent with the prediction of network pharmacology. IκB, p65 and COX-2 were identified as key targets and this study demonstrated for the first time that JE reverses 2.5% DSS-induced UC in mice via the IκB/p65/COX-2/arachidonic acid pathway. This study reveals the complex mechanisms underlying the therapeutic effects of JE on UC and provides a new approach to identifying the underlying mechanisms of the pharmacological action of Chinese natural medicines such as JE.
Collapse
Affiliation(s)
- Jinyan Qiu
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Guanlin Xiao
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Minjuan Yang
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xuejun Huang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Dake Cai
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Canhui Xie
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhao Chen
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Xiaoli Bi
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China.
| | - Aili Xu
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China.
| |
Collapse
|
2
|
Wu F, Zhang X, Zhang S, Zhang Y, Feng Y, Jiang Z, Shi Y, Zhang S, Tu W. Construction of an immune-related lncRNA-miRNA-mRNA regulatory network in radiation-induced esophageal injury in rats. Int Immunopharmacol 2023; 122:110606. [PMID: 37423154 DOI: 10.1016/j.intimp.2023.110606] [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: 04/19/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
Radiation-induced esophageal injury (RIEI) is an adverse reaction of radiation therapy in patients with esophageal cancer, lung cancer and other malignant tumors. Competitive endogenous RNA (ceRNA) network is known to play a significant role in the onset and progression of many diseases, but the exact mechanism of ceRNA in RIEI has not been fully elucidated. In this study, rat esophaguses were obtained after conducting irradiation under different doses (0 Gy, 25 Gy, 35 Gy). Total RNA was extracted and mRNA, lncRNA, circRNA, and miRNA sequencing was performed. Multiple dose-dependent differentially expressed RNAs (dd-DERs), including 870 lncRNAs, 82 miRNAs, 2478 mRNAs, were obtained through the integration of differential expression analysis and dose-dependent screening (35 Gy ≥ 25 Gy > 0 Gy, or 35 Gy ≤ 25 Gy < 0 Gy). Co-expression analysis and prediction of the binding site in dd-DER were conducted and 27 lncRNAs, 20 miRNAs, and 168 mRNAs were selected to construct a ceRNA network. As the immune microenvironment is crucial for RIEI progression, we constructed an immune-related ceRNA network consisting of 11 lncRNAs, 9 miRNAs, and 9 mRNAs. The expression levels of these immune-related RNAs were verified by RT-qPCR. Immune infiltration analysis showed that the RNAs in the immune-related ceRNA network were mainly associated with the proportion of monocytes, M2 macrophages, activated NK cells, and activated CD4+ memory T cells. Drug sensitivity analysis was conducted based on the expression levels of mRNAs in the immune-related ceRNA network, and small molecule drugs with preventive and therapeutic effects on RIEI were identified. In summary, an immune-related ceRNA network associated with RIEI progression was constructed in this study. The findings provide useful information on new potential targets for the prevention and treatment of RIEI.
Collapse
Affiliation(s)
- Fengping Wu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Xiaolin Zhang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Shuaijun Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China; Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuehua Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China; Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yahui Feng
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China; NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China
| | - Zhiqiang Jiang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Yuhong Shi
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Shuyu Zhang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China; Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China; Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China; NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China.
| | - Wenling Tu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China; School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, China; NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China.
| |
Collapse
|
3
|
Liu M, Sun Z, Tang Y, Zhang S, Luo J. The Regulation of Exosome-Mediated miR-132-3p/miR-132-3p-UUU on Radiation-Induced Esophageal Injury. Radiat Res 2023; 200:151-161. [PMID: 37327123 DOI: 10.1667/rade-22-00070.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/23/2023] [Indexed: 06/18/2023]
Abstract
Radiation-induced esophageal injury (RIEI) is a major dose-limiting complication of radiotherapy, mainly acute esophagitis. However, understanding of radiation injury and repair mechanisms in esophageal epithelial cells remains limited. MiR-132-3p and its uridylated isoform (miR-132-3p-UUU) are upregulated in radiation esophageal injury, yet their role in radiation-induced esophageal injury progression remains unexplored. We expressed miR-132-3p and its uridine form in irradiated human esophageal epithelial cells (HEEC) and secreted exosomes was examined by real-time polymerase chain reaction (RT-PCR). Cell proliferation, migration, apoptosis and colony formation were used to determine biological effects. Cell cycle assays and dual luciferase reporter assays were used to assess the relationship between miR-132-3p and its uridylated isoforms and MEF2A. The addition of miR-132-3p mimics or overexpression of miR-132-3p significantly inhibited the proliferation and migration of esophageal epithelial cells (HEEC cells as well as primary cells) and increased radiation damage. This was reversed by its uridylated isoform by reducing binding to MEF2A and regulating the cell cycle. Furthermore, miR-132-3p and its triuridylated isomer also regulate apoptosis after irradiation through pathways other than reactive oxygen species (ROS). In conclusion, our data reveal that radiation-induced miR-132-3p uridylation and exosome-mediated intercellular communication and tri-uridylated isoforms are protective against radiation-induced esophageal injury. Furthermore, miR-132-3p offers new opportunities as a promising biomarker widely present in human body fluids for the prediction of radiation esophagitis as a biomarker.
Collapse
Affiliation(s)
- Muzi Liu
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yiting Tang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
| | - Shuyu Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
4
|
Yao J, Zhang J, Wang J, Lai Q, Yuan W, Liu Z, Cheng S, Feng Y, Jiang Z, Shi Y, Jiang S, Tu W. Transcriptome Profiling Unveils a Critical Role of IL-17 Signaling-Mediated Inflammation in Radiation-Induced Esophageal Injury in Rats. Dose Response 2022; 20:15593258221104609. [PMID: 35677348 PMCID: PMC9168911 DOI: 10.1177/15593258221104609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023] Open
Abstract
Elucidation of the molecular mechanisms involving the initiation and progression
of radiation-induced esophageal injury (RIEI) is important for prevention and
treatment. Despite ongoing advances, the underlying mechanisms controlling RIEI
remain largely unknown. In the present study, RNA-seq was performed to
characterize mRNA profiles of the irradiated rat esophagus exposed to 0, 25, or
35 Gy irradiation. Bioinformatics analyses including dose-dependent
differentially expressed genes (DEGs), Gene Ontology (GO), Kyoto Encyclopedia of
Gene and Genome (KEGG) pathway, protein-protein interaction (PPI) network, and
immune infiltration were performed. 134 DEGs were screened out with a
dose-dependent manner (35 Gy > 25 Gy > control, or 35 Gy < 25 Gy <
control). GO and KEGG analyses showed that the most significant mechanism was
IL-17 signaling-mediated inflammatory response. 5 hub genes, Ccl11, Cxcl3,
Il17a, S100a8, and S100a9, were identified through the intersection of the DEGs
involved in inflammatory response, IL-17 pathway, and PPI network. Additionally,
immune infiltration analysis showed the activation of macrophages, monocytes, T
cells, NKT cells, and neutrophils, among which macrophages, monocytes, and
neutrophils might be the main sources of S100a8 and S100a9. Thus, these findings
further our understanding on the molecular biology of RIEI and may help develop
more effective therapeutic strategies.
Collapse
Affiliation(s)
- Jia Yao
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jinkang Zhang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Jinlong Wang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Qian Lai
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Weijun Yuan
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Zhongyu Liu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuanghua Cheng
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Yahui Feng
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Zhiqiang Jiang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Yuhong Shi
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Sheng Jiang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Wenling Tu
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|