1
|
Li X, Zhang Z, Li C, Liu J, Fang Q, Zhang M, Huang J. Novel applications of metformin in the treatment of septic myocardial injury based on metabolomics and network pharmacology. Eur J Pharmacol 2025; 986:177141. [PMID: 39566813 DOI: 10.1016/j.ejphar.2024.177141] [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: 08/14/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND While metformin has shown promise in treating septic myocardial injury (SMI), its underlying mechanisms and impact on metabolic disturbances remain poorly understood. METHODS This study employed an integrated approach of metabolomics and network pharmacology to identify key targets and pathways through which metformin may act against SMI. Findings were validated using a lipopolysaccharide (LPS)-induced mouse model. RESULTS Metformin was found to counter myocardial metabolic disruptions, indicated by the reversal of 49 metabolites primarily involved in purine metabolism, pantothenate and CoA biosynthesis, and histidine metabolism. In vivo, metformin significantly improved survival rates and cardiac function, reduced cardiomyocyte apoptosis, and inhibited inflammation and oxidative stress in LPS-induced mice. Integrated analyses identified 27 potential targets for metformin in SMI treatment. KEGG pathway analysis revealed significant enrichment in TNF, HIF-1, IL-17, and PI3K/AKT signaling pathways, while protein-protein interaction analysis pinpointed ten core targets, including IL6, IL1B, CCL2, CASP3, MMP9, HIF1A, IGF1, NOS3, MMP2, and LEP. Molecular docking and dynamics simulations demonstrated metformin's high affinity for these core targets. Further, RT-qPCR and Western blot analyses confirmed that metformin modulates core target expression to mitigate SMI. Notably, our data underscore the importance of PI3K/AKT and MMP2/MMP9 signaling pathways in SMI therapy. CONCLUSION This study elucidates the metabolic and molecular mechanisms of metformin in SMI treatment, supporting its potential repurposing for SMI.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zihan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chaohong Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Jun Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghua Fang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muzi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Zaheen A, Rajkhowa S, Al‐Hussain SA, Zaki MEA. Integrated computational strategies for Polypharmacological profiling and identification of anti-inflammatory targets in Rungia pectinata L. J Cell Mol Med 2024; 28:e70158. [PMID: 39629503 PMCID: PMC11615512 DOI: 10.1111/jcmm.70158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 12/08/2024] Open
Abstract
Rungia pectinata L. is an ethnomedicinal herb belonging to the Acanthaceae family and it presents a promising avenue for medicinal exploration, deeply rooted in traditional practices. Earlier research has demonstrated that the herb can effectively relieve the classic symptoms of inflammation. Nevertheless, comprehensive studies into the mechanisms underlying R. pectinata's beneficial impact on inflammation pathways, remain scarce. Hence, we employed an integrated approach combining network pharmacology, molecular docking and molecular dynamics simulations to explore the mechanisms underlying R. pectinata's anti-inflammatory activity. For this study, seven inflammation-related active ingredients were identified among 38 candidates, revealing 22 intersecting genes associated with inflammation. Protein-protein interaction (PPI) networks revealed three therapeutic targets: IL1B, PTGS2 and SRC. GO and KEGG pathway enrichment analyses indicated that the effects of R. pectinata are mediated by genes related to inflammation and cancer. Molecular docking studies identified trans-nerolidyl formate and widdrol as lead compounds while molecular dynamics simulations indicated stable compound-target complexes, with MM-PBSA calculations showing superior free energy values for SRC, suggesting implications in cancer pathways. Overall, this study offers valuable insights into the anti-inflammatory effects of R. pectinata, which may be mediated through key pathways involved in inflammation and cancer. This highlights the potential of R. pectinata in both anti-inflammatory and anticancer therapies. However, further experimental validation is necessary to confirm these findings.
Collapse
Affiliation(s)
- Alaiha Zaheen
- Centre for Biotechnology and BioinformaticsDibrugarh UniversityDibrugarhIndia
| | - Sanchaita Rajkhowa
- Centre for Biotechnology and BioinformaticsDibrugarh UniversityDibrugarhIndia
| | - Sami A. Al‐Hussain
- Department of ChemistryImam Mohammad Ibn Saud Islamic University (IMSIU)RiyadhSaudi Arabia
| | - Magdi E. A. Zaki
- Department of ChemistryImam Mohammad Ibn Saud Islamic University (IMSIU)RiyadhSaudi Arabia
| |
Collapse
|
3
|
Hao Z, Lu C, Wang M, Li S, Wang Y, Yan Y, Ding Y, Li Y. Systematic investigation on the pharmaceutical components and mechanism of the treatment against zebrafish enteritis by Sporisorium reilianum f. sp. reilianum based on histomorphology and pathology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118574. [PMID: 39019416 DOI: 10.1016/j.jep.2024.118574] [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: 03/23/2024] [Revised: 06/21/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sporisorium reilianum f. sp. reilianum (SSR) is a fungus isolated from a medicinal plant. Recorded in the "Compilation of National Chinese Herbal Medicine" and "Compendium of Materia Medica," it was used for preventing and treating intestinal diseases, enhancing immune function, etc. In this study, we investigated the chemical composition and bioactivity of SSR. Network pharmacology is utilized for predictive analysis and targeting pathway studies of anti-inflammatory bowel disease (IBD) mechanisms. Pharmacological activity against enteritis is evaluated using zebrafish (Danio rerio) as model animals. AIM OF THE STUDY To reveal the treatment of IBD by SSR used as traditional medicine and food, based on molecular biology identification of SSR firstly, and the pharmaceutical components & its toxicities, biological activity & mechanism of SSR were explored. MATERIALS AND METHODS Using chromatography and zebrafish IBD model induced by dextran sulfate sodium (DSS), nine compounds were first identified by nuclear magnetic resonance (NMR). The toxicity of ethanol crude extract and monomers from SSR were evaluated by evaluating the phenotypic characteristics of zebrafish embryos and larvae, histomorphology and pathology of the zebrafish model guided by network pharmacology were conducted. RESULTS The zebrafish embryo development did not show toxicity. The molecular docking and enrichment pathway results predicted that metabolites 3 & 4 (N-trans- feruloyl-3-methoxytyramine & N-cis-feruloyl-3-methoxytyramine) and 7 & 8 (4-N- trans-p-coumaroyltyramine & 4-N-cis--p-coumaroyltyramine) have anti-enteritis activities. This paper lays an experimental foundation for developing new drugs and functional foods.
Collapse
Affiliation(s)
- Zezhuang Hao
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Chang Lu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Mengtong Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Shuxia Li
- Jinmanwu Agricultural Science and Technology Development Co., LTD., Liaoyuan, 136200, China.
| | - Ye Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Yuli Yan
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Yuling Ding
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Yong Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
4
|
Zhu X, Wang B, Yu H, Li C, Zhao Y, Zhong Y, Tang W, Zhou Y, Huang X, Zhu H, Wu Y, Yang K, Wei Y, Gao Z, Dong J. Icariin attenuates asthmatic airway inflammation via modulating alveolar macrophage activation based on network pharmacology and in vivo experiments. J Gene Med 2024; 26:e3718. [PMID: 38979822 DOI: 10.1002/jgm.3718] [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: 09/07/2023] [Revised: 03/23/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Icariin (ICA) inhibits inflammatory response in various diseases, but the mechanism underlying ICA treating airway inflammation in asthma needs further understood. We aimed to predict and validate the potential targets of ICA against asthma-associated airway inflammation using network pharmacology and experiments. METHODS The ovalbumin-induced asthma-associated airway inflammation mice model was established. The effects of ICA were evaluated by behavioral, airway hyperresponsiveness, lung pathological changes, inflammatory cell and cytokines counts. Next, the corresponding targets of ICA were mined via the SEA, CTD, HERB, PharmMapper, Symmap database and the literature. Pubmed-Gene and GeneCards databases were used to screen asthma and airway inflammation-related targets. The overlapping targets were used to build an interaction network, analyze gene ontology and enrich pathways. Subsequently, flow cytometry, quantitative real-time PCR and western blotting were employed for validation. RESULTS ICA alleviated the airway inflammation of asthma; 402 targets of ICA, 5136 targets of asthma and 4531 targets of airway inflammation were screened; 216 overlapping targets were matched and predicted ICA possesses the potential to modulate asthmatic airway inflammation by macrophage activation/polarization. Additionally, ICA decreased M1 but elevated M2. Potential targets that were disrupted by asthma inflammation were restored by ICA treatment. CONCLUSIONS ICA alleviates airway inflammation in asthma by inhibiting the M1 polarization of alveolar macrophages, which is related to metabolic reprogramming. Jun, Jak2, Syk, Tnf, Aldh2, Aldh9a1, Nos1, Nos2 and Nos3 represent potential targets of therapeutic intervention. The present study enhances understanding of the anti-airway inflammation effects of ICA, especially in asthma.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Bin Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Congcong Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuanyuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Huahe Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yueren Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Kai Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhen Gao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Zhong T, Chen S, Deng K, Guan J, Zhang J, Lu F, Shichen M, Lv R, Liu Z, Liu Y, Chang P, Liu Z. Magnesium alleviates extracellular histone-induced apoptosis and defective bacterial phagocytosis in macrophages by regulating intracellular calcium signal. Int Immunopharmacol 2024; 132:111870. [PMID: 38547771 DOI: 10.1016/j.intimp.2024.111870] [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: 01/04/2024] [Revised: 02/18/2024] [Accepted: 03/12/2024] [Indexed: 05/01/2024]
Abstract
Extracellular histones have been determined as important mediators of sepsis, which induce excessive inflammatory responses in macrophages and impair innate immunity. Magnesium (Mg2+), one of the essential nutrients of the human body, contributes to the proper regulation of immune function. However, no reports indicate whether extracellular histones affect survival and bacterial phagocytosis in macrophages and whether Mg2+ is protective against histone-induced macrophage damage. Our clinical data revealed a negative correlation between circulating histone and monocyte levels in septic patients, and in vitro experiments confirmed that histones induced mitochondria-associated apoptosis and defective bacterial phagocytosis in macrophages. Interestingly, our clinical data also indicated an association between lower serum Mg2+ levels and reduced monocyte levels in septic patients. Moreover, in vitro experiments demonstrated that Mg2+ attenuated histone-induced apoptosis and defective bacterial phagocytosis in macrophages through the PLC/IP3R/STIM-mediated calcium signaling pathway. Importantly, further animal experiments proved that Mg2+ significantly improved survival and attenuated histone-mediated lung injury and macrophage damage in histone-stimulated mice. Additionally, in a cecal ligation and puncture (CLP) + histone-induced injury mouse model, Mg2+ inhibited histone-mediated apoptosis and defective phagocytosis in macrophages and further reduced bacterial load. Overall, these results suggest that Mg2+ supplementation may be a promising treatment for extracellular histone-mediated macrophage damage in sepsis.
Collapse
Affiliation(s)
- Tao Zhong
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sainan Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ke Deng
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianbin Guan
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaqi Zhang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Furong Lu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Maoyou Shichen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ronggui Lv
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhifeng Liu
- Department of Medicine Intensive Care Units, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China.
| | - Yong Liu
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhanguo Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Geng Q, Liu B, Fan D, Cao Z, Li L, Lu P, Lin L, Yan L, Xiong Y, He X, Lu J, Chen P, Lu C. Strictosamide ameliorates LPS-induced acute lung injury by targeting ERK2 and mediating NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117593. [PMID: 38113987 DOI: 10.1016/j.jep.2023.117593] [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: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) ranks among the deadliest pulmonary diseases, significantly impacting mortality and morbidity. Presently, the primary treatment for ALI involves supportive therapy; however, its efficacy remains unsatisfactory. Strictosamide (STR), an indole alkaloid found in the Chinese herbal medicine Nauclea officinalis (Pierre ex Pit.) Merr. & Chun (Wutan), has been found to exhibit numerous pharmacological properties, particularly anti-inflammatory effects. AIM OF THE STUDY This study aimes to systematically identify and validate the specific binding proteins targeted by STR and elucidate its anti-inflammatory mechanism in lipopolysaccharide (LPS)-induced ALI. MATERIALS AND METHODS Biotin chemical modification, protein microarray analysis and network pharmacology were conducted to screen for potential STR-binding proteins. The binding affinity was assessed through surface plasmon resonance (SPR), cellular thermal shift assay (CETSA) and molecular docking, and the anti-inflammatory mechanism of STR in ALI treatment was assessed through in vivo and in vitro experiments. RESULTS Biotin chemical modification, protein microarray and network pharmacology identified extracellular-signal-regulated kinase 2 (ERK2) as the most important binding proteins among 276 candidate STR-interacting proteins and nuclear factor-kappaB (NF-κB) pathway was one of the main inflammatory signal transduction pathways. Using SPR, CETSA, and molecular docking, we confirmed STR's affinity for ERK2. In vitro and in vivo experiments demonstrated that STR mitigated inflammation by targeting ERK2 to modulate the NF-κB signaling pathway in LPS-induced ALI. CONCLUSIONS Our findings indicate that STR can inhibit the NF-κB signaling pathway to attenuate LPS-induced inflammation by targeting ERK2 and decreasing phosphorylation of ERK2, which could be a novel strategy for treating ALI.
Collapse
Affiliation(s)
- Qi Geng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Danping Fan
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China; Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Beijing, 100700, PR China
| | - Zhiwen Cao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Peipei Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Lin Lin
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Lan Yan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Yibai Xiong
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Peng Chen
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China; Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Beijing, 100700, PR China.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China.
| |
Collapse
|
7
|
Alipourfard I, Darvishi M, Khalighfard A, Ghazi F, Mobed A. Nanomaterial-based methods for sepsis management. Enzyme Microb Technol 2024; 174:110380. [PMID: 38147783 DOI: 10.1016/j.enzmictec.2023.110380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/26/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023]
Abstract
Sepsis is a serious disease caused by an impaired host immune response to infection, resulting in organ dysfunction, tissue damage and is responsible for high in-hospital mortality (approximately 20%). Recently, WHO documented sepsis as a global health priority. Nevertheless, there is still no effective and specific therapy for clinically detecting sepsis. Nanomaterial-based approaches have appeared as promising tools for identifying bacterial infections. In this review, recent biosensors are introduced and summarized as nanomaterial-based platforms for sepsis management and severe complications. Biosensors can be used as tools for the diagnosis and treatment of sepsis and as nanocarriers for drug delivery. In general, diagnostic methods for sepsis-associated bacteria, biosensors developed for this purpose are presented in detail, and their strengths and weaknesses are discussed. In other words, readers of this article will gain a comprehensive understanding of biosensors and their applications in sepsis management.
Collapse
Affiliation(s)
- Iraj Alipourfard
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia, Katowice, Poland
| | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Arghavan Khalighfard
- Department of Nursing and Midwifery٫ Faculty of Midwifery٬ Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farhood Ghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5154853431, Iran
| | - Ahmad Mobed
- Infectious and Tropical Diseases Research Center, Clinical Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Zhang W. Critical roles of S100A12, MMP9, and PRTN3 in sepsis diagnosis: Insights from multiple microarray data analyses. Comput Biol Med 2024; 171:108222. [PMID: 38447501 DOI: 10.1016/j.compbiomed.2024.108222] [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: 11/12/2023] [Revised: 02/16/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Sepsis, characterized by systemic inflammatory response syndrome and life-threatening organ dysfunction, remains a significant global cause of disability and death. Despite its impact, reliable biomarkers for sepsis diagnosis are yet to be identified. OBJECTIVE This study aims to investigate and identify key genes and pathways in sepsis through the analysis of multiple microarray datasets, providing potential treatment targets for future clinical trials. METHODS Two independent gene expression profiles (GSE54514 and GSE69528) were downloaded from the Gene Expression Omnibus (GEO) database. After merging and batch normalization, differentially expressed genes (DEGs) were obtained using the "limma" package. Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) were performed using "R" software. A Protein-Protein Interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes (STRING). The top 10 hub genes were identified using Cytoscape. A Nomogram model for predicting sepsis occurrence was constructed and evaluated. RESULTS Bioinformatic analysis of 210 sepsis and 91 control blood samples identified 72 DEGs. GO analyses revealed associations with immune response processes. GSEA indicated involvement in key signaling pathways. S100A12, MMP9, and PRTN3 were identified as independent risk factors for sepsis. CONCLUSION This study unveils critical genes and pathways in sepsis through bioinformatic methods. S100A12, MMP9, and PRTN3 may play essential roles in the immune response to infection, influencing sepsis prognosis.
Collapse
Affiliation(s)
- Wenyuan Zhang
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
9
|
Yi B, Lv F, Zhang N, Lin J, Xu K, Li C, Li P, Zhao M. Exploring the pharmacological mechanisms of Biyan Qingdu Granula in the treatment after nasopharyngeal carcinoma radiotherapy based on UPLC/Q-TOF MS, network pharmacology and molecular docking. J Pharm Biomed Anal 2024; 239:115830. [PMID: 38096633 DOI: 10.1016/j.jpba.2023.115830] [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: 07/21/2023] [Revised: 09/30/2023] [Accepted: 10/27/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Biyan Qingdu Granula (BYQD) is a traditional Chinese medicine (TCM) formula commonly used for post-radiotherapy treatment of nasopharyngeal carcinoma (NPC). Despite its extensive use, the underlying pharmacological mechanisms have yet to be fully elucidated. METHODS UPLC/Q-TOF MS was used to comprehensively analyze the chemical composition of BYQD. Additionally, an everted gut sac model, coupled with UPLC/Q-TOF MS, was used to screen and identify the active ingredients. Subsequently, we conducted a network pharmacological analysis to delve into the potential mechanisms of these active ingredients. Molecular docking experiments were also performed to assess the interactions between active ingredients and potential core targets. RESULTS The findings revealed the identification of 62 identical ingredients upon comparing the sample solution and intestinal absorbed solution of BYQD. We constructed a protein-protein interaction (PPI) network, which led to the identification of five core targets, namely, TP53, STAT3, MAPK1, SRC and AKT1. Through the construction of a drug-active ingredient-intersection target network, we identified Quercetin, Luteolin, Eupatilin, Magnoflorine, Acacetin and other compound as potential active ingredients. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis suggested that pathways in cancer, PI3K-Akt signaling pathway, lipid and atherosclerosis, proteoglycans in cancer, and the MAPK signaling pathway might play the key roles in the treatment of NPC after radiotherapy using BYQD. Molecular docking results corroborated strong binding activity between the putative core targets and the corresponding key active ingredients. CONCLUSION This study provides a preliminary revelation of the active ingredients and potential pharmacological mechanisms of BYQD in the post-radiotherapy treatment of NPC. These findings establish a vital theoretical basis and serve as a scientific reference for the future investigating the pharmacological mechanisms and clinical application of BYQD.
Collapse
Affiliation(s)
- Bojiao Yi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fengyi Lv
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, Shandong, China
| | - Na Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Juan Lin
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Keyi Xu
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Chuyuan Li
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Peng Li
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China.
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
10
|
Zhong T, Zhang J, Chen S, Chen S, Deng K, Guan J, Yang J, Lv R, Liu Z, Liu Y, Chang P, Liu Z. MAGNESIUM SULFATE AMELIORATES HISTONE-INDUCED COAGULATION DYSFUNCTION AND LUNG DAMAGE IN MICE. Shock 2024; 61:132-141. [PMID: 37988072 DOI: 10.1097/shk.0000000000002263] [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: 11/22/2023]
Abstract
ABSTRACT Introduction: Extracellular histones have been determined as significant mediators of sepsis, which can induce endothelial cell injury and promote coagulation activation, and ultimately contribute to multiorgan failure. Evidence suggests that magnesium sulfate (MgSO 4 ) exerts a potential coagulation-modulating activity; however, whether MgSO 4 ameliorates histone-induced coagulation dysfunction and organ damage remains unclear. Methods: To measure circulating histone levels, blood specimens were collected from septic patients and mice, and the relationship between circulating histone levels, coagulation parameters, and Mg 2+ levels in sepsis was investigated. Furthermore, to explore the possible protective effects of MgSO 4 , we established a histone-induced coagulation model in mice by intravenous histone injection. The survival rate of mice was assessed, and the histopathological damage of the lungs (including endothelial cell injury and coagulation status) was evaluated using various methods, including hematoxylin and eosin staining, immunohistochemistry, immunofluorescence, electron microscopy, and quantitative polymerase chain reaction. Results: The circulating histone levels in septic patients and mice were significantly associated with several coagulation parameters. In septic patients, histone levels correlated negatively with platelet counts and positively with prothrombin time and D-dimer levels. Similarly, in cecal ligation and puncture mice, histones correlated negatively with platelet counts and positively with D-dimer levels. Interestingly, we also observed a positive link between histones and Mg 2+ levels, suggesting that Mg 2+ with anticoagulant activity is involved in histone-mediated coagulation alterations in sepsis. Further animal experiments confirmed that MgSO 4 administration significantly improved survival and attenuated histone-mediated endothelial cell injury, coagulation dysfunction, and lung damage in mice. Conclusion: These results suggest that therapeutic targeting of histone-mediated endothelial cell injury, coagulation dysfunction, and lung damage, for example, with MgSO 4 , may be protective in septic individuals with elevated circulating histone levels.
Collapse
Affiliation(s)
- Tao Zhong
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaqi Zhang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shanjia Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sainan Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ke Deng
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianbin Guan
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingjing Yang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ronggui Lv
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhifeng Liu
- Department of Medicine Intensive Care Units, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China
| | - Yong Liu
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhanguo Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Liu J, Cao C, Jin Y, Wang Y, Ma X, Li J, Guo S, Yang J, Niu J, Liang X. Induced neural stem cells suppressed neuroinflammation by inhibiting the microglial pyroptotic pathway in intracerebral hemorrhage rats. iScience 2023; 26:107022. [PMID: 37360683 PMCID: PMC10285565 DOI: 10.1016/j.isci.2023.107022] [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: 01/11/2023] [Revised: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Intracerebral hemorrhage usually manifests as strong neuroinflammation and neurological deficits. There is an urgent need to explore effective methods for the treatment of intracerebral hemorrhage. The therapeutic effect and the possible mechanism of induced neural stem cell transplantation in an intracerebral hemorrhage rat model are still unclear. Our results showed that transplantation of induced neural stem cells could improve neurological deficits by inhibiting inflammation in an intracerebral hemorrhage rat model. Additionally, induced neural stem cell treatment could effectively suppress microglial pyroptosis, which might occur through inhibiting the NF-κB signaling pathway. Induced neural stem cells could also regulate the polarization of microglia and promote the transition of microglia from pro-inflammatory phenotypes to anti-inflammatory phenotypes to exert their anti-inflammatory effects. Overall, induced neural stem cells may be a promising tool for the treatment of intracerebral hemorrhage and other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Jiaxin Liu
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Chuanshang Cao
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Yiran Jin
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Yan Wang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Xiaona Ma
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Jiahui Li
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Songlin Guo
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Jiancheng Yang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 750004 Yinchuan, China
| | - Xueyun Liang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, 750001 Yinchuan, China
| |
Collapse
|
12
|
Xu HY, Li QC, Zhou WJ, Zhang HB, Chen ZX, Peng N, Gong SY, Liu B, Zeng F. Anti-Oxidative and Anti-Aging Effects of Probiotic Fermented Ginseng by Modulating Gut Microbiota and Metabolites in Caenorhabditis elegans. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2023:10.1007/s11130-023-01055-9. [PMID: 36947370 DOI: 10.1007/s11130-023-01055-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Antioxidative and antiaging abilities of probiotic fermented ginseng (PG) were evaluated in Caenorhabditis elegans (C. elegans). Lifespan and effect on heat stress and acute oxidative stress in C. elegans were significantly enhanced by PG. Antioxidative enzymes such as T-SOD, GSH-PX, CAT were significantly up-regulated, and MDA, ROS and apoptosis levels were significantly down-regulated. At the same time, PG exerted antioxidant and anti-aging activities by reducing the expression of DAF-2 mRNA and increasing the expression of SKN-1 and SOD-3 mRNA in C. elegans. In addition, the mechanism of antioxidative and antiaging activities of PG was explored through gut microbiota sequencing and untargeted metabolomics. The results of gut microbiota indicated that PG could significantly improve the composition and structure of microbes in the gut of C. elegans, and the relative abundance of beneficial bacteria was up-regulated. Untargeted metabolomic results elucidated that PG modulated antioxidant and antiaging activities through neuroactive ligand-receptor interaction, Citrate cycle (TCA cycle), pyruvate metabolism, ascorbate and aldarate metabolism and D-Arginine and D-ornithine metabolism of C. elegans. These results indicated that PG had excellent antioxidant and anti-aging activities, providing research value for the development of functional foods and improvement of aging-related diseases.
Collapse
Affiliation(s)
- Huan-Yi Xu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
| | - Quan-Cen Li
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
| | - Wen-Jie Zhou
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
| | - Hai-Bo Zhang
- The Hubei Provincial Key Laboratory of Yeast Function, Yichang, 443003, China
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Zhi-Xian Chen
- The Hubei Provincial Key Laboratory of Yeast Function, Yichang, 443003, China
| | - Ning Peng
- The Hubei Provincial Key Laboratory of Yeast Function, Yichang, 443003, China
| | - Shi-Yu Gong
- The Hubei Provincial Key Laboratory of Yeast Function, Yichang, 443003, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
| | - Feng Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China.
| |
Collapse
|
13
|
Zhang D, Wang L, Wang Z, Shi X, Tang W, Jiang L, Bo Ran Yi BYCH, Lv X, Hu C, Xiao D. Immunological responses of septic rats to combination therapy with thymosin α1 and vitamin C. Open Life Sci 2023; 18:20220551. [PMID: 36816800 PMCID: PMC9922062 DOI: 10.1515/biol-2022-0551] [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: 07/02/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 02/10/2023] Open
Abstract
This study investigated the effect of combined thymosin α1 and vitamin C (Tα1 + VitC) on the immunological responses of septic rats. Five groups were designed. The septic model was established by the cecal ligation puncture (CLP) method. The sham group did not undergo CLP, the model group was given normal saline solution, the Tα1 group was given Tα1 (200 µg/kg), the VitC group was given VitC (200 mg/kg), and the Tα1 + VitC group was given Tα1 + VitC. Specimens for immunological analyses were collected at 6, 12, 24, and 48 h posttreatment in each group except for the sham group (only at 48 h). CD4 + CD25 + T cells in the peripheral blood and dendritic cell (DC) proportions in the spleen were analyzed by flow cytometry. Tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), transforming growth factor-β (TGF-ß1), and nuclear factor kappa-B (NF-κB) were measured by ELISA. CD4 + CD25 + T cells and OX62 + DCs levels significantly increased in the model group and decreased in the Tα1 and/or VitC treatment groups. Similarly, the levels of TNF-α, IL-6, TGF-ß1, and NF-κB significantly increased in the model group and decreased in the Tα1, VitC, and Tα1 + VitC groups, indicating that combined Tα1 and VitC therapy may help regulate the immunological state of patients with sepsis, thereby improving prognosis.
Collapse
Affiliation(s)
- Daquan Zhang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Lu Wang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Zhigao Wang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Xiaohui Shi
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Wen Tang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Long Jiang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Ba Yin Cha Han Bo Ran Yi
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Xinwei Lv
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Congyu Hu
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Dong Xiao
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| |
Collapse
|
14
|
Pan W, Jie W, Huang H. Vascular calcification: Molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e200. [PMID: 36620697 PMCID: PMC9811665 DOI: 10.1002/mco2.200] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
Vascular calcification (VC) is recognized as a pathological vascular disorder associated with various diseases, such as atherosclerosis, hypertension, aortic valve stenosis, coronary artery disease, diabetes mellitus, as well as chronic kidney disease. Therefore, it is a life-threatening state for human health. There were several studies targeting mechanisms of VC that revealed the importance of vascular smooth muscle cells transdifferentiating, phosphorous and calcium milieu, as well as matrix vesicles on the progress of VC. However, the underlying molecular mechanisms of VC need to be elucidated. Though there is no acknowledged effective therapeutic strategy to reverse or cure VC clinically, recent evidence has proved that VC is not a passive irreversible comorbidity but an active process regulated by many factors. Some available approaches targeting the underlying molecular mechanism provide promising prospects for the therapy of VC. This review aims to summarize the novel findings on molecular mechanisms and therapeutic interventions of VC, including the role of inflammatory responses, endoplasmic reticulum stress, mitochondrial dysfunction, iron homeostasis, metabolic imbalance, and some related signaling pathways on VC progression. We also conclude some recent studies on controversial interventions in the clinical practice of VC, such as calcium channel blockers, renin-angiotensin system inhibitions, statins, bisphosphonates, denosumab, vitamins, and ion conditioning agents.
Collapse
Affiliation(s)
- Wei Pan
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Wei Jie
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
15
|
He Z, Gao K, Dong L, Liu L, Qu X, Zou Z, Wu Y, Bu D, Guo JC, Zhao Y. Drug screening and biomarker gene investigation in cancer therapy through the human transcriptional regulatory network. Comput Struct Biotechnol J 2023; 21:1557-1572. [PMID: 36879883 PMCID: PMC9984461 DOI: 10.1016/j.csbj.2023.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/19/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
A complex and vast biological network regulates all biological functions in the human body in a sophisticated manner, and abnormalities in this network can lead to disease and even cancer. The construction of a high-quality human molecular interaction network is possible with the development of experimental techniques that facilitate the interpretation of the mechanisms of drug treatment for cancer. We collected 11 molecular interaction databases based on experimental sources and constructed a human protein-protein interaction (PPI) network and a human transcriptional regulatory network (HTRN). A random walk-based graph embedding method was used to calculate the diffusion profiles of drugs and cancers, and a pipeline was constructed by using five similarity comparison metrics combined with a rank aggregation algorithm, which can be implemented for drug screening and biomarker gene prediction. Taking NSCLC as an example, curcumin was identified as a potentially promising anticancer drug from 5450 natural small molecules, and combined with differentially expressed genes, survival analysis, and topological ranking, we obtained BIRC5 (survivin), which is both a biomarker for NSCLC and a key target for curcumin. Finally, the binding mode of curcumin and survivin was explored using molecular docking. This work has a guiding significance for antitumor drug screening and the identification of tumor markers.
Collapse
Affiliation(s)
- Zihao He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinchi Qu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhengkai Zou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Wu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Dechao Bu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Jin-Cheng Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.,Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
16
|
Investigating Key Targets of Dajianzhong Decoction for Treating Crohn’s Disease Using Weighted Gene Co-Expression Network. Processes (Basel) 2022. [DOI: 10.3390/pr11010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: Crohn’s disease (CD) is an inflammatory bowel disease, cases of which have substantially increased in recent years. The classical formula Dajianzhong decoction (DD, Japanese: Daikenchuto) is often used to treat CD, but few studies have evaluated related therapeutic mechanisms. In this study, we investigated the potential targets and mechanisms of DD used for treating CD at the molecular level through the weighted gene co-expression network. Methods: The main chemical components of the three DD herbs (Zanthoxylum bungeanum Maxim., Zingiber officinale (Willd.) Rosc., and Ginseng Radix et Rhizoma) were searched for using the HERB database. The targets for each component were identified using the SwissTargetPrediction and HERB databases, whereas the disease targets for CD were retrieved from the GeneCards and DisGeNET databases. The functional enrichment analysis was performed on the common targets of DD and CD. High-throughput sequencing data for CD patients were retrieved from the Gene Expression Omnibus database, and WGCNA was performed to identify the key targets. The association between the key targets and DD ingredients was verified using molecular docking. Results: By analyzing the interaction targets between DD and CD, 196 overlapping genes were identified. The enrichment results indicated that the PI3K-AKT, TNF, MAPK, and IL-17 signaling pathways influenced the mechanism of action of DD in counteracting CD. Combined with WGCNA, four differentially expressed genes (SLC6A4, NOS2, SHBG, and ABCB1) and their corresponding 24 compounds were closely related to the occurrence of CD. Conclusions: By integrating gene co-expression network analysis, this study preliminarily reveals the internal molecular mechanism of DD in treating CD from a systematic perspective, validated by molecular docking. However, these findings require further validation.
Collapse
|
17
|
Xia T, Liang X, Liu CS, Hu YN, Luo ZY, Tan XM. Network Pharmacology Integrated with Transcriptomics Analysis Reveals Ermiao Wan Alleviates Atopic Dermatitis via Suppressing MAPK and Activating the EGFR/AKT Signaling. Drug Des Devel Ther 2022; 16:4325-4341. [PMID: 36578822 PMCID: PMC9790806 DOI: 10.2147/dddt.s384927] [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: 08/10/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background Ermiao Wan (EMW) is commonly used to treat atopic dermatitis (AD) in China. However, the pharmacological mechanisms underlying the action of EMW against AD remain unclear. Purpose We aimed to determine the mechanisms underlying the effectiveness of EMW in the treatment of AD. Methods We evaluated the effect of EMW on AD induced by dinitrochlorobenzene (DNCB) in BALB/C mice. To clarify the key components of EMW in AD treatment, the main components of EMW were identified using HPLC. Serum pharmacochemistry was used to analyze the absorbed ingredients from blood. Based on the phytochemical results, network pharmacology and molecular docking were used to predict the action of EMW. Skin transcriptomic analysis was used to validate the network pharmacology results. RT-qPCR,ELISA, and immunohistochemical were performed to validate the results of skin transcriptomics. Results EMW improved the symptoms of AD, with less rashes, less spontaneous scratching, less inflammatory cell infiltration, and fewer allergic reactions. The established HPLC method is simple and reliable. Chlorogenic acid, phellodendrine, magnoflorine, jatrorrhizine, palmatine, berberine, and atractylodin were the key effective ingredients with a high blood concentration. Fifty-seven primary causal targets of EMW against AD were identified. These targets are mainly involved in ErbB signaling pathways including EGFR, AKT1, MAPK8, JUN, MAPK1. Molecular docking showed that EGFR, AKT1, MAPK8, JUN, MAPK1 had good binding force with EMW. In AD mice, EMW regulated the EGFR/AKT signaling through upregulation of Grb2, GAB1, Raf-1, EGFR, and AKT, and downregulation of MAPK1 and JUN, compared to that in the MD group. Conclusion EMW could alleviate AD through activating EGFR/AKT signaling and suppressing MAPK. This study provides a theoretical basis for the clinical use of EMW.
Collapse
Affiliation(s)
- Ting Xia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Xiao Liang
- School of Pharmaceutical Sciences, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Chang-Shun Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Yan-Nan Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China,Correspondence: Xiao-Mei Tan, Tel/Fax + 86-020-61648265, Email
| |
Collapse
|
18
|
Xu H, You M, Xiang X, Zhao J, Yuan P, Chu L, Xie C. Molecular Mechanism of Epimedium Extract against Ischemic Stroke Based on Network Pharmacology and Experimental Validation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3858314. [PMID: 36338345 PMCID: PMC9633197 DOI: 10.1155/2022/3858314] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2024]
Abstract
Ischemic stroke exhibits high morbidity, disability, and mortality, and treatments for ischemic stroke are limited despite intensive research. The potent neuroprotective benefits of Epimedium against ischemic stroke have gained lots of interest. Nevertheless, systematic research on the direct role and mechanisms of Epimedium in ischemic stroke is still lacking. Network pharmacology analysis coupled with experimental verification was utilized to systematically evaluate the potential pharmacological mechanism of Epimedium against ischemic stroke. The TCMSP database was used to mine the bioactive ingredients and Epimedium's targets. The DrugBank, OMIM, and GeneCards databases were employed to identify potential targets of ischemic stroke. GO and KEGG pathway analyses were also carried out. The interaction between active components and hub targets was confirmed via molecular docking. An experimental ischemic stroke model was used to evaluate the possible therapeutic mechanism of Epimedium. As a result, 23 bioactive compounds of Epimedium were selected, and 30 hub targets of Epimedium in its function against ischemic stroke were identified, and molecular docking results demonstrated good binding. The IL-17 signaling pathway was revealed as a potentially significant pathway, with the NF-κB and MAPK/ERK signaling pathways being involved. Furthermore, in vivo experiments demonstrated that Epimedium treatment could improve neurological function and reduce infarct volume. Additionally, Epimedium reduced the activation of microglia and astrocytes in both the ischemic penumbra of the hippocampus and cerebral cortex following ischemic stroke. Western blot and RT-qPCR analyses demonstrated that Epimedium not only depressed the expression of IL-1β, TNF-α, IL-6, and IL-4 but also inhibited the NF-κB and MAPK/ERK signaling pathways. This study applied network pharmacology and in vivo experiment to explore possible mechanism of Epimedium's role against ischemic stroke, which provides insight into the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongbei Xu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Mingyao You
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Xiang Xiang
- Neurosurgery Department of Chongqing University, Three Gorges Hospital, Chongqing 400010, China
| | - Jun Zhao
- Department of Neurosurgery, Dazhou Hospital of Integrated Traditional and Western Medicine, 635000, China
| | - Ping Yuan
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Lan Chu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, China
| | - Chenchen Xie
- Department of Neurology, Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu 610081, China
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
19
|
Xi R, Wan Y, Yang L, Zhang J, Yang L, Yang S, Chai R, Mu F, Sun Q, Yan R, Wu Z, Li S. Investigating Celastrol's Anti-DCM Targets and Mechanisms via Network Pharmacology and Experimental Validation. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7382130. [PMID: 35845929 PMCID: PMC9278495 DOI: 10.1155/2022/7382130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022]
Abstract
Methods Data from TCMSP and GEO databases were utilized to identify targets for Celastrol on DCM. The relationship between the major targets and conventional glycolipid metabolism was obtained with Spearman correlation analysis. Experiments on animals were conducted utilizing healthy control (HC), low-dose Celastrol interventions (CL), and no intervention groups (NC), all of which had 8 SD rats in each group. To study alterations in signaling molecules, RT-PCR was performed. Results There were 76 common targets and 5 major targets for Celastrol-DCM. Celastrol have been found to regulate AGE-RAGE, TNF, MAPK, TOLL-like receptors, insulin resistance, and other signaling pathways, and they are closely linked to adipocytokines, fatty acid metabolism, glycolipid biosynthesis, and glycosylphosphati-dylinositol biosynthesis on DCM. These five major targets have been found to regulate these pathways. Experiments on rats indicated that P38 MAPK was considerably elevated in the cardiac tissue from rats in the CL and NC groups compared to the HC group, and the difference was statistically significant (P < 0.01). Significant differences were seen between the CL and NC groups in P38 MAPK levels, with a statistical significance level of less than 0.05. Conclusion Celastrol may play a role in reversing energy remodeling, anti-inflammation, and oxidative stress via modulating p38 protein expression in the MAPK pathway, which have been shown in the treatment of DCM.
Collapse
Affiliation(s)
- Rui Xi
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongxin Wan
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lihong Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jingying Zhang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Liu Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shuai Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Chai
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fengchen Mu
- Department of Vascular Medicine, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, China
| | - Qiting Sun
- Department of Nuclear Medicine, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, China
| | - Rui Yan
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
20
|
Ming T, Dong M, Song X, Li X, Kong Q, Fang Q, Wang J, Wu X, Xia Z. Integrated Analysis of Gene Co-Expression Network and Prediction Model Indicates Immune-Related Roles of the Identified Biomarkers in Sepsis and Sepsis-Induced Acute Respiratory Distress Syndrome. Front Immunol 2022; 13:897390. [PMID: 35844622 PMCID: PMC9281548 DOI: 10.3389/fimmu.2022.897390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a series of clinical syndromes caused by immunological response to severe infection. As the most important and common complication of sepsis, acute respiratory distress syndrome (ARDS) is associated with poor outcomes and high medical expenses. However, well-described studies of analysis-based researches, especially related bioinformatics analysis on revealing specific targets and underlying molecular mechanisms of sepsis and sepsis-induced ARDS (sepsis/se-ARDS), still remain limited and delayed despite the era of data-driven medicine. In this report, weight gene co-expression network based on data from a public database was constructed to identify the key modules and screen the hub genes. Functional annotation by enrichment analysis of the modular genes also demonstrated the key biological processes and signaling pathway; among which, extensive immune-involved enrichment was remarkably associated with sepsis/se-ARDS. Based on the differential expression analysis, least absolute shrink and selection operator, and multivariable logistic regression analysis of the screened hub genes, SIGLEC9, TSPO, CKS1B and PTTG3P were identified as the candidate biomarkers for the further analysis. Accordingly, a four-gene-based model for diagnostic prediction assessment was established and then developed by sepsis/se-ARDS risk nomogram, whose efficiency was verified by calibration curves and decision curve analyses. In addition, various machine learning algorithms were also applied to develop extra models based on the four genes. Receiver operating characteristic curve analysis proved the great diagnostic and predictive performance of these models, and the multivariable logistic regression of the model was still found to be the best as further verified again by the internal test, training, and external validation cohorts. During the development of sepsis/se-ARDS, the expressions of the identified biomarkers including SIGLEC9, TSPO, CKS1B and PTTG3P were all regulated remarkably and generally exhibited notable correlations with the stages of sepsis/se-ARDS. Moreover, the expression levels of these four genes were substantially correlated during sepsis/se-ARDS. Analysis of immune infiltration showed that multiple immune cells, neutrophils and monocytes in particular, might be closely involved in the process of sepsis/se-ARDS. Besides, SIGLEC9, TSPO, CKS1B and PTTG3P were considerably correlated with the infiltration of various immune cells including neutrophils and monocytes during sepsis/se-ARDS. The discovery of relevant gene co-expression network and immune signatures might provide novel insights into the pathophysiology of sepsis/se-ARDS.
Collapse
Affiliation(s)
- Tingqian Ming
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Mingyou Dong
- College of Medical Laboratory Science, Youjiang Medical College for Nationalities, Baise, China
| | - Xuemin Song
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xingqiao Li
- School of Computer, Wuhan University, Wuhan, China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Qing Fang
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, China
- *Correspondence: Zhongyuan Xia, ; Xiaojing Wu,
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, China
- *Correspondence: Zhongyuan Xia, ; Xiaojing Wu,
| |
Collapse
|
21
|
Nag A, Verma P, Paul S, Kundu R. In Silico Analysis of the Apoptotic and HPV Inhibitory Roles of Some Selected Phytochemicals Detected from the Rhizomes of Greater Cardamom. Appl Biochem Biotechnol 2022; 194:4867-4891. [PMID: 35670907 PMCID: PMC9171093 DOI: 10.1007/s12010-022-04006-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 11/29/2022]
Abstract
Occurrence of cervical cancer, caused due to persistent human papilloma virus (HPV) infection, is common in women of developing countries. As the conventional treatments are expensive and associated with severe side effects, there is a need to find safer alternatives, which is affordable and less toxic to the healthy human cells. Present study aimed to evaluate the anti-HPV and apoptotic potential of four compounds from the greater cardamom (Amomum subulatum Roxb. var. Golsey), namely rhein, phytosphingosine, n-hexadecenoic acid and coronarin E. Their anti-HPV and apoptotic potential were studied against viral E6, E7 and few anti-apoptotic proteins of host cell (BCL2, XIAP, LIVIN) by in silico docking technique. Phytochemicals from the plant extract were analysed and identified by LC/MS and GC/MS. Involvement of the target proteins in various biological pathways was determined through KEGG. Structural optimization of the three-dimensional structures of the ligands (four phytochemicals and control drug) was done by Avogadro1.1. Receptor protein models were built using ProMod3 and other advanced tools. Pharmacophore modelling of the selected phytochemicals was performed in ZINCPharmer. Swiss ADME studies were undertaken to determine drug likeness. The ligands and proteins were digitally docked in DockThor docking program. Protein flexibility-molecular dynamic simulation helped to study protein–ligand stability in real time. Finally, the correlation of evaluated molecules was studied by the use of principal component analysis (PCA) based on the docking scores. All the ligands were found to possess apoptotic and anti-cancer activities and did not violate Lipinsky criteria. n-Hexadecanoic acid and its analogues showed maximum efficacy against the target proteins. All the protein–ligand interactions were found to be stable. The uncommon phytochemicals identified from rhizomes of greater cardamom have anti-cancer, apoptotic and HPV inhibitory potentials as analysed by docking and other in silico studies, which can be utilized in drug development after proper experimental validation.
Collapse
Affiliation(s)
- Anish Nag
- Department of Life Sciences, CHRIST (Deemed to Be University), Bangalore, India
| | - Preeti Verma
- Laboratory of Advanced Cell Biology, Department of Botany, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, India, 700019
| | - Subhabrata Paul
- Institute of Health Sciences, Presidency University (2Nd Campus), Newtown, Kolkata, India
| | - Rita Kundu
- Laboratory of Advanced Cell Biology, Department of Botany, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, India, 700019.
| |
Collapse
|
22
|
Qin Q, Qin L, Xie R, Peng S, Guo C, Yang B. Insight Into Biological Targets and Molecular Mechanisms in the Treatment of Arsenic-Related Dermatitis With Vitamin A via Integrated in silico Approach. Front Nutr 2022; 9:847320. [PMID: 35685889 PMCID: PMC9171494 DOI: 10.3389/fnut.2022.847320] [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: 01/02/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to arsenic (As), an inorganic poison, may lead to skin lesions, including dermatitis. Vitamin A (VA), a fat-soluble vitamin essential for mucous membrane integrity, plays a key role in skin protection. Although the beneficial actions of VA are known, the anti-As-related dermatitis effects of VA action remain unclear. Hence, in this study, we aimed to interpret and identify the core target genes and therapeutic mechanisms of VA action in the treatment of As-related dermatitis through integrated in silico approaches of network pharmacology and molecular docking. We integrated the key VA-biological target-signaling pathway-As-related dermatitis networks for identifying core drug targets and interaction pathways associated with VA action. The network pharmacology data indicated that VA may possess potential activity for treating As-related dermatitis through the effective regulation of core target genes. An enrichment analysis in biological processes further revealed multiple immunoregulation-associated functions, including interferon-gamma production and negative regulation of T-cell activation and production of molecular mediator of immune response. An enrichment analysis in molecular pathways mainly uncovered multiple biological signaling, including natural killer cell mediated cytotoxicity, autophagy, apoptosis, necroptosis, platelet activation involved in cell fate, and immunity regulations. Molecular docking study was used to identify docked well core target proteins with VA, including Jun, tumor protein p53 (TP53), mitogen-activated protein kinase-3 (MAPK3), MAPK1, and MAPK14. In conclusion, the potential use of VA may suppress the inflammatory stress and enhance the immunity against As-related dermatitis. In the future, VA might be useful in the treatment of dermatitis associated with As through multi-targets and multi-pathways in clinical practice.
Collapse
Affiliation(s)
- Qiuhai Qin
- Department of Surgery, The People’s Hospital of Gangbei District, Guigang, China
| | - Lixiu Qin
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Ruitang Xie
- Department of Surgery, The People’s Hospital of Gangbei District, Guigang, China
| | - Shuihua Peng
- Department of Pharmacy, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Chao Guo
- Department of Pharmacy, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
- *Correspondence: Chao Guo,
| | - Bin Yang
- College of Pharmacy, Guangxi Medical University, Nanning, China
- Bin Yang,
| |
Collapse
|
23
|
Li R, Huang X, Yang L, Liang X, Huang W, Lai KP, Zhou L. Integrated Analysis Reveals the Targets and Mechanisms in Immunosuppressive Effect of Mesalazine on Ulcerative Colitis. Front Nutr 2022; 9:867692. [PMID: 35662946 PMCID: PMC9161553 DOI: 10.3389/fnut.2022.867692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/15/2022] [Indexed: 12/21/2022] Open
Abstract
Background Ulcerative colitis (UC) is an inflammatory bowel disease that causes inflammation and ulcers in the digestive tract. Approximately 3 million US adults suffer from this disease. Mesalazine, an anti-inflammatory agent, is commonly used for the treatment of UC. However, some studies have demonstrated side effects of mesalazine, such as acute pancreatitis and hypereosinophilia. Therefore, a better understanding of the anti-inflammatory mechanism of mesalazine in UC could help improve the effectiveness of the drug and reduce its side effects. In this study, we used a dextran sodium sulfate-induced UC mouse model, and applied network pharmacology and omics bioinformatics approaches to uncover the potential pharmaceutical targets and the anti-inflammatory mechanism of mesalazine. Results Network pharmacology analysis identified the core targets of mesalazine, biological processes, and cell signaling related to immunity and inflammatory responses mediated by mesalazine. Molecular docking analysis then indicated possible binding motifs on the core targets (including TNF-α, PTGS2, IL-1β, and EGFR). Metabolomics and 16S metagenomic analyses highlighted the correlation between gut microbiota and metabolite changes caused by mesalazine in the UC model. Conclusions Collectively, the omics and bioinformatics approaches and the experimental data unveiled the detailed molecular mechanisms of mesalazine in UC treatment, functional regulation of the gut immune system, and reduction of intestinal inflammation. More importantly, the identified core targets could be targeted for the treatment of UC.
Collapse
Affiliation(s)
- Rong Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Xue Huang
- Department of Gastroenterology, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Lu Yang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Xiao Liang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Wenjun Huang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- *Correspondence: Wenjun Huang
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- Keng Po Lai
| | - Liming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- Liming Zhou ;
| |
Collapse
|
24
|
Xu H, Qin L, Nie L, Li L, Guo P, Chen Y, Huang C, Su M, Yang B. Biotargets for mediation of arsenic–induced coronary heart disease by calycosin. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2053947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hongyuan Xu
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Lixiu Qin
- College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, PR People’s Republic of China
| | - Litao Nie
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR People’s Republic of China
| | - Lin Li
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Peng Guo
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Yizhao Chen
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Chuang Huang
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Min Su
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR People’s Republic of China
| | - Bin Yang
- College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, PR People’s Republic of China
| |
Collapse
|
25
|
Song J, Zhang H, Wang ZX, Wang J. The antioxidant activity, α-glucosidase and acetylcholinesterase inhibition activity, and chemical composition of Paeonia delavayi petal. FOOD QUALITY AND SAFETY 2022. [DOI: 10.1093/fqsafe/fyac020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Abstract
Objectives
This study aimed to evaluate the functional activity and phytochemical compositions in the flower petals of Paeonia delavayi with different colors.
Materials and Methods
P. delavayi petal extracts were prepared by maceration in methanol, including purple petal extract (PPE), red petal extract (RPE) and yellow petal extract (YPE), and their antioxidant activity, α-glucosidase and acetylcholinesterase inhibition activities were evaluated. To correlate these measured activities to phytochemicals in the petals, UPLC-MS/MS-based metabolomics method was applied to profile the compositions in the petals of different colors. Finally, the KEGG metabolic pathways database was used to identify the related metabolic pathways that are responsible for the production of these polyphenolic phytochemicals in the petals.
Results
The results showed that PPE had the highest total phenolic content (TPC), total flavonoid content (TFC), and the strongest ABTS· + scavenging ability, ferric reducing antioxidant power, and acetylcholinesterase inhibition ability in all three samples, while YPE showed the strongest DPPH· scavenging activity and α-glucosidase inhibition ability. A total of 232 metabolites were detected in the metabolomic analysis, 198 of which were flavonoids, chalcones, flavonols, and anthocyanins. Correlation analysis indicated that Peonidin-3-O-arabinoside and cyanidin-3-O-arabinoside were the major contributors to their antioxidant activity. Principal component analysis showed a clear separation between these three petals. In addition, a total of 38, 98, and 96 differential metabolites were identified in PPE, RPE, and YPE, respectively. Pathway enrichment revealed 6 KEGG pathways displayed significant enrichment differences, of which the anthocyanin biosynthesis, flavone and flavonol biosynthesis were the most enriched signaling pathways. It revealed the potential reason for the differences in metabolic and functional levels between different colors of P. delavayi petals.
Conclusions
P. delavayi petals of different colors have different metabolite contents and functional activities, of which the anthocyanin, flavone, and flavonol metabolites are critical in its functional activities, suggesting the anthocyanin biosynthesis, flavone and flavonol biosynthesis pathways be the key pathways responsible for both the petal color and bioactive phytochemicals in P. delavayi flowers.
Collapse
|
26
|
Wu J, Wang Z, Xu S, Fu Y, Gao Y, Wu Z, Yu Y, Yuan Y, Zhou L, Li P. Analysis of the role and mechanism of EGCG in septic cardiomyopathy based on network pharmacology. PeerJ 2022; 10:e12994. [PMID: 35287352 PMCID: PMC8917800 DOI: 10.7717/peerj.12994] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/02/2022] [Indexed: 01/11/2023] Open
Abstract
Background Septic cardiomyopathy (SC) is a common complication of sepsis that leads to an increase in mortality. The pathogenesis of septic cardiomyopathy is unclear, and there is currently no effective treatment. EGCG (epigallocatechin gallate) is a polyphenol that has anti-inflammatory, antiapoptotic, and antioxidative stress effects. However, the role of EGCG in septic cardiomyopathy is unknown. Methods Network pharmacology was used to predict the potential targets and molecular mechanisms of EGCG in the treatment of septic cardiomyopathy, including the construction and analysis of protein-protein interaction (PPI) network, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and molecular docking. The mouse model of septic cardiomyopathy was established after intraperitoneal injection of LPS (lipopolysaccharide). The myocardial protective effect of EGCG on septic mice is observed by cardiac ultrasound and HE staining. RT-PCR is used to verify the expression level of the EGCG target in the septic cardiomyopathy mouse model. Results A total of 128 anti-SC potential targets of EGCGareselected for analysis. The GO enrichment analysis and KEGG pathway analysis results indicated that the anti-SC targets of EGCG mainly participate in inflammatory and apoptosis processes. Molecular docking results suggest that EGCG has a high affinity for the crystal structure of six targets (IL-6 (interleukin-6), TNF (tumor necrosis factor), Caspase3, MAPK3 (Mitogen-activated protein kinase 3), AKT1, and VEGFA (vascular endothelial growth factor)), and the experimental verification result showed levated expression of these 6 hub targets in the LPS group, but there is an obvious decrease in expression in the LPS + EGCG group. The functional and morphological changes found by echocardiography and HE staining show that EGCG can effectively improve the cardiac function that is reduced by LPS. Conclusion Our results reveal that EGCG may be a potentially effective drug to improve septic cardiomyopathy. The potential mechanism by which EGCG improves myocardial injury in septic cardiomyopathy is through anti-inflammatory and anti-apoptotic effects. The anti-inflammatory and anti-apoptotic effects of EGCG occur not only through direct binding to six target proteins (IL-6,TNF-α, Caspase3, MAPK3, AKT1, and VEGFA) but also by reducing their expression.
Collapse
Affiliation(s)
- Ji Wu
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Zhenhua Wang
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Shanling Xu
- Department of Cardiovascular, Medicine, Fuzhou First People’s Hospital, Fu Zhou, China
| | - Yang Fu
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Yi Gao
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Zuxiang Wu
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Yun Yu
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Yougen Yuan
- Department of Cardiovascular, The Three Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Lin Zhou
- Department of Cardiovascular, The Three Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Ping Li
- Department of Cardiovascular, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| |
Collapse
|
27
|
Xu H, Qin J, Qin L, Guo C, Yang B. Bioinformatics and In Silico Findings Uncover Bio-Targets of Calycosin Against Heart Failure and Diabetes Mellitus. Front Endocrinol (Lausanne) 2022; 13:790619. [PMID: 35898453 PMCID: PMC9309256 DOI: 10.3389/fendo.2022.790619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/23/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Heart failure (HF) and diabetes mellitus (DM) are life-threatening diseases. However, existing clinical drugs to treat HF complicated with DM are relatively limited. In this study, we performed a viable bioinformatics strategy combining network pharmacology and molecular docking to identify potential anti-HF and -DM targets and therapeutic mechanisms of calycosin, a functional phytoestrogen. METHODS Web-based databases were used to collect candidate genes/targets of calycosin and HF/DM and then identify the hub bio-targets of calycosin against HF/DM. Using the online-available database, all functional processes and signaling pathways of calycosin against HF/DM were screened and identified before further visualization. RESULTS All potential bio-targets of calycosin and HF/DM were collected, and 20 hub targets of calycosin against HF/DM were identified. Interestingly, molecular docking findings indicated that mitogen-activated protein kinase-1 (MAPK1), β-arrestin 1 (ARRB1), and homologue-1 (ABL1) may be potent pharmacological targets of calycosin against HF/DM. In addition, all primary molecular functions of calycosin against HF/DM were identified, including regulating protein binding, ubiquitination, and the metabolic process. Furthermore, the top molecular pathways of calycosin against HF/DM were revealed, including cardiomyocyte and chemokine signaling pathways. CONCLUSION Our bioinformatics analysis uncovered the network targets and therapeutic mechanisms of calycosin against HF/DM. For the first time, the current in silico findings revealed that the identified hub targets may be used to screen and treat HF/DM.
Collapse
Affiliation(s)
- Hongyuan Xu
- Cardiology Department, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Jingru Qin
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Lixiu Qin
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Chao Guo
- Department of Pharmacy, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
- *Correspondence: Chao Guo, ; Bin Yang,
| | - Bin Yang
- College of Pharmacy, Guangxi Medical University, Nanning, China
- *Correspondence: Chao Guo, ; Bin Yang,
| |
Collapse
|
28
|
Hasankhani A, Bahrami A, Sheybani N, Aria B, Hemati B, Fatehi F, Ghaem Maghami Farahani H, Javanmard G, Rezaee M, Kastelic JP, Barkema HW. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front Immunol 2021; 12:789317. [PMID: 34975885 PMCID: PMC8714803 DOI: 10.3389/fimmu.2021.789317] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background The recent emergence of COVID-19, rapid worldwide spread, and incomplete knowledge of molecular mechanisms underlying SARS-CoV-2 infection have limited development of therapeutic strategies. Our objective was to systematically investigate molecular regulatory mechanisms of COVID-19, using a combination of high throughput RNA-sequencing-based transcriptomics and systems biology approaches. Methods RNA-Seq data from peripheral blood mononuclear cells (PBMCs) of healthy persons, mild and severe 17 COVID-19 patients were analyzed to generate a gene expression matrix. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules in healthy samples as a reference set. For differential co-expression network analysis, module preservation and module-trait relationships approaches were used to identify key modules. Then, protein-protein interaction (PPI) networks, based on co-expressed hub genes, were constructed to identify hub genes/TFs with the highest information transfer (hub-high traffic genes) within candidate modules. Results Based on differential co-expression network analysis, connectivity patterns and network density, 72% (15 of 21) of modules identified in healthy samples were altered by SARS-CoV-2 infection. Therefore, SARS-CoV-2 caused systemic perturbations in host biological gene networks. In functional enrichment analysis, among 15 non-preserved modules and two significant highly-correlated modules (identified by MTRs), 9 modules were directly related to the host immune response and COVID-19 immunopathogenesis. Intriguingly, systemic investigation of SARS-CoV-2 infection identified signaling pathways and key genes/proteins associated with COVID-19's main hallmarks, e.g., cytokine storm, respiratory distress syndrome (ARDS), acute lung injury (ALI), lymphopenia, coagulation disorders, thrombosis, and pregnancy complications, as well as comorbidities associated with COVID-19, e.g., asthma, diabetic complications, cardiovascular diseases (CVDs), liver disorders and acute kidney injury (AKI). Topological analysis with betweenness centrality (BC) identified 290 hub-high traffic genes, central in both co-expression and PPI networks. We also identified several transcriptional regulatory factors, including NFKB1, HIF1A, AHR, and TP53, with important immunoregulatory roles in SARS-CoV-2 infection. Moreover, several hub-high traffic genes, including IL6, IL1B, IL10, TNF, SOCS1, SOCS3, ICAM1, PTEN, RHOA, GDI2, SUMO1, CASP1, IRAK3, HSPA5, ADRB2, PRF1, GZMB, OASL, CCL5, HSP90AA1, HSPD1, IFNG, MAPK1, RAB5A, and TNFRSF1A had the highest rates of information transfer in 9 candidate modules and central roles in COVID-19 immunopathogenesis. Conclusion This study provides comprehensive information on molecular mechanisms of SARS-CoV-2-host interactions and identifies several hub-high traffic genes as promising therapeutic targets for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Behzad Aria
- Department of Physical Education and Sports Science, School of Psychology and Educational Sciences, Yazd University, Yazd, Iran
| | - Behzad Hemati
- Biotechnology Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahsa Rezaee
- Department of Medical Mycology, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
29
|
Yang L, Yang J, Liang X, Huang W, Zhang X, Li R. Uncovering antiobesity-related hypertension targets and mechanisms of metformin, an antidiabetic medication. Bioengineered 2021; 12:4757-4767. [PMID: 34334083 PMCID: PMC8806643 DOI: 10.1080/21655979.2021.1954581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/07/2021] [Indexed: 01/10/2023] Open
Abstract
Metformin, a common clinical drug used to treat diabetes mellitus, is found with potential antiobese actions as reported in increasing evidences. However, the detailed mechanisms of metformin-antiobesity-related hypertension remain unrevealed. We have utilized the bioinformatics strategy, including network pharmacology and molecular docking analyses, to uncover pharmacological targets and molecular pathways of bioactive compounds against clinical disorders, such as cancers, coronavirus disease 2019. In this report, the in-silico approaches using network pharmacology and molecular docking was utilized to identify the core targets, pharmacological functions and mechanisms of metformin against obesity-related hypertension. The networking analysis identified 154 differentially expressed genes of obesity and hypertension, and 21 interaction genes, 6 core genes of metformin treating obesity-related hypertension. As results, molecular docking findings indicated the binding capability of metformin with key proteins, including interleukin 6 (IL-6) and chemokine (C-C motif) Ligand 2 (CCL2) expressed in obesity- and hypertension-dependent tissues. Metformin-exerted antihypertension/obesity actions involved in metabolic regulation, inflammatory suppression. And antihypertension/obesity mechanisms of metformin were revealed, including regulation of inflammatory and immunological signaling pathways for ameliorating microenvironmental homeostasis in targeting tissues. In conclusion, our current bioinformatics findings have uncovered all pharmacological targets, biological functions and signaling pathways of metformin treating obesity-related hypertension, thus promoting its clinical application in future.
Collapse
Affiliation(s)
- Lu Yang
- Faculty of Basic Medicine, Guilin Medical University, Guilin, PR China
| | - Jianxin Yang
- Cardiology Department Area 1, Guigang City People’s Hospital, the Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, PR China
| | - Xiao Liang
- Faculty of Basic Medicine, Guilin Medical University, Guilin, PR China
| | - Wenjun Huang
- Laboratory of Environmental Pollutants and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Xiaoxi Zhang
- Faculty of Basic Medicine, Guilin Medical University, Guilin, PR China
| | - Rong Li
- Laboratory of Environmental Pollutants and Integrative Omics, Guilin Medical University, Guilin, PR China
| |
Collapse
|
30
|
Qin L, Huang D, Huang J, Qin F, Huang H. Integrated Analysis and Finding Reveal Anti-Liver Cancer Targets and Mechanisms of Pachyman ( Poria cocos Polysaccharides). Front Pharmacol 2021; 12:742349. [PMID: 34603055 PMCID: PMC8484528 DOI: 10.3389/fphar.2021.742349] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
This bioinformatics study aimed to characterize and certify crucial anti-cancer targets, functional processes, and molecular mechanisms of Pachyman in treating hepatocellular carcinoma (HCC) by using pharmacology network and molecular docking analyses, by experimental validation. The crucial anti-HCC targets of Pachyman, including ALB, VEGFA, TNF, CASP3, SRC, EGF, CXCR4, STAT3, HRAS, HSP90AA1, MMP9, BCL2L1, FGF2, and PTPRC, were identified. In addition, the correlative networks of all crucial biotargets of Pachyman in treating HCC were created accordingly. Functionally, these crucial genes were correlated using angiogenesis and neoplastic metastasis of HCC. Interestingly, the molecular docking findings indicated that ALB and VEGFA in HCC might be potent pharmacological targets of Pachyman. In experimental validation, the clinical samples of HCC showed reduced ALB protein expression and increased VEGFA protein level. Following Pachyman treatments in vitro, the intracellular level of ALB protein was elevated, whereas the cellular content of VEGFA protein was downregulated. Taken together, current bioinformatics findings based on pharmacology network and molecular docking analyses elucidate the detailed molecular targets and signaling mechanisms of Pachyman in treating HCC. Interestingly, validated biotargets of ALB and VEGFA may be main potential biomarkers for detecting HCC medically.
Collapse
Affiliation(s)
- Li Qin
- Department of Oncology, Liuzhou Worker's Hospital, Liuzhou, China
| | - Dongning Huang
- Department of Oncology, Liuzhou Worker's Hospital, Liuzhou, China
| | - Jian Huang
- Department of Oncology, Liuzhou Worker's Hospital, Liuzhou, China
| | - Fuhui Qin
- Department of Oncology, Liuzhou Worker's Hospital, Liuzhou, China
| | - Haixin Huang
- Department of Oncology, Liuzhou Worker's Hospital, Liuzhou, China
| |
Collapse
|
31
|
Abstract
The aim of current study was to exhume the potential targets and molecular mechanisms of oxyresveratrol, a structurally re-constructed resveratrol, for treating liver cancer through bioinformatics investigation and experimentative validation. To start with, the network pharmacology approach and molecular docking technology were used to uncover all candidate targets of oxyresveratrol to treat liver cancer, accompanied with identified anti-liver cancer targets including estrogen receptor 1 (ESR1), epidermal growth factor receptor (EGFR). In addition, more pharmacological mechanisms of oxyresveratrol against liver cancer were revealed in details. In experimental verification, the clinical samples of liver cancer showed elevated ESR1, EGFR mRNA expressions. The in-vitro data indicated that intracellular contents of ESR1, EGFR mRNAs in oxyresveratrol-treated liver cancer cells were reduced. Taken together, the bioinformatics and validated findings have highlighted detailed pharmacological targets and molecular mechanisms of oxyresveratrol for treating liver cancer. Following with experimental verification, the identified genes of ESR1, EGFR may function as potential screening anti-liver cancer markers.
Collapse
|
32
|
He J, Xu W, Zheng X, Zhao B, Ni T, Yu P, Deng S, Pan X, Chen E, Mao E, Bian X. Vitamin C reduces vancomycin-related nephrotoxicity through the inhibition of oxidative stress, apoptosis, and inflammation in mice. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1319. [PMID: 34532456 PMCID: PMC8422136 DOI: 10.21037/atm-21-3294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022]
Abstract
Background Vancomycin (VCM) is an antibiotic widely used to treat a range of serious bacterial infections; however, it is associated with nephrotoxicity. Vitamin C (VC) is a classical antioxidant that can alleviate various organ injuries and inflammatory responses by reducing inflammation and oxidative stress. This study aimed to examine the effect of VC on VCM-related nephrotoxicity in mice. Methods Mice were randomized into four groups: control, VCM (400 mg/kg/day), VCM (400 mg/kg/day) + VC (200 mg/kg/day), and VC (200 mg/kg/day) groups. Both VCM and VC were administered via intraperitoneal injection for 7 d, after which kidney and blood samples were collected and evaluated. Creatinine (Cr), blood urea nitrogen (BUN), superoxide dismutase (SOD), malondialdehyde (MDA), interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and nuclear factor-κB (NF-κB) were measured. Results In the VCM group, kidney index, renal injury score, cell apoptosis, serum Cr and BUN, and kidney Cr, BUN, MDA, IL-1β, IL-6, TNF-α, and NF-κB were higher compared to the control group (all P<0.05), while body weight and kidney SOD activity were lower (both P<0.05). By contrast, no differences were observed between the control and VC groups (VC and VCM + VC groups) for all these indicators. Conclusions The antioxidant VC reduces VCM-related renal injury by reducing oxidative stress, cell apoptosis, and inflammation.
Collapse
Affiliation(s)
- Juan He
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyun Xu
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxiao Zheng
- Department of Pharmacy, Xuzhou First People's Hospital, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bing Zhao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tongtian Ni
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Yu
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyu Deng
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxia Pan
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolan Bian
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Ouyang Y, Rong Y, Wang Y, Guo Y, Shan L, Yu X, Li L, Si J, Li X, Ma K. A Systematic Study of the Mechanism of Acacetin Against Sepsis Based on Network Pharmacology and Experimental Validation. Front Pharmacol 2021; 12:683645. [PMID: 34483900 PMCID: PMC8415621 DOI: 10.3389/fphar.2021.683645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
Sepsis is a dysregulated systemic response to infection, and no effective treatment options are available. Acacetin is a natural flavonoid found in various plants, including Sparganii rhizoma, Sargentodoxa cuneata and Patrinia scabiosifolia. Studies have revealed that acacetin potentially exerts anti-inflammatory and antioxidative effects on sepsis. In this study, we investigated the potential protective effect of acacetin on sepsis and revealed the underlying mechanisms using a network pharmacology approach coupled with experimental validation and molecular docking. First, we found that acacetin significantly suppressed pathological damage and pro-inflammatory cytokine expression in mice with LPS-induced fulminant hepatic failure and acute lung injury, and in vitro experiments further confirmed that acacetin attenuated LPS-induced M1 polarization. Then, network pharmacology screening revealed EGFR, PTGS2, SRC and ESR1 as the top four overlapping targets in a PPI network, and GO and KEGG analyses revealed the top 20 enriched biological processes and signalling pathways associated with the therapeutic effects of acacetin on sepsis. Further network pharmacological analysis indicated that gap junctions may be highly involved in the protective effects of acacetin on sepsis. Finally, molecular docking verified that acacetin bound to the active sites of the four targets predicted by network pharmacology, and in vitro experiments further confirmed that acacetin significantly inhibited the upregulation of p-src induced by LPS and attenuated LPS-induced M1 polarization through gap junctions. Taken together, our results indicate that acacetin may protect against sepsis via a mechanism involving multiple targets and pathways and that gap junctions may be highly involved in this process.
Collapse
Affiliation(s)
- Yuanshuo Ouyang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Yi Rong
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Yanming Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Yanli Guo
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Liya Shan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Xiushi Yu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Li Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
| | - Junqiang Si
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| |
Collapse
|
34
|
Al Sulaiman K, Aljuhani O, Saleh KB, Badreldin HA, Al Harthi A, Alenazi M, Alharbi A, Algarni R, Al Harbi S, Alhammad AM, Vishwakarma R, Aldekhyl S. Ascorbic acid as an adjunctive therapy in critically ill patients with COVID-19: a propensity score matched study. Sci Rep 2021; 11:17648. [PMID: 34480041 PMCID: PMC8417267 DOI: 10.1038/s41598-021-96703-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
Ascorbic acid represents an appealing option for clinicians to utilize in the context of the global COVID-19 pandemic due to its proposed clinical efficacy, relative safety, and low cost. The aim of this study was to evaluate the efficacy and safety of using ascorbic acid in supplemental doses as adjunctive therapy for patients critically ill with COVID-19. This was a two-center, non-interventional, retrospective cohort study. All critically ill adult patients admitted to ICU with a confirmed COVID-19 diagnosis between March 1st and December 31st, 2020, were included in the final analysis. The study was conducted at two large governmental tertiary hospitals in Saudi Arabia. The purpose was to investigate the clinical outcomes of low-dose ascorbic acid as adjunctive therapy in COVID-19 after propensity score matching using baseline severity scores, systematic use of corticosteroids, and study centers. A number of 739 patients were included in this study, among whom 296 patients were included after propensity score matching. There was no association between the administration of ascorbic acid and in-hospital mortality or the 30-day mortality [OR (95% CI) 0.77 (0.47, 1.23), p value = 0.27 and OR (95% CI) 0.73 (0.43, 1.20), p value = 0.21, respectively]. Using ascorbic acid was associated with a lower incidence of thrombosis compared with the non-ascorbic-acid group [6.1% vs. 13% respectively; OR (95% CI) 0.42 (0.184, 0.937), p value = 0.03]. Low dose of ascorbic acid as an adjunctive therapy in COVID-19 critically ill patients was not associated with mortality benefits, but it was associated with a lower incidence of thrombosis. Further studies are required to confirm these findings.
Collapse
Affiliation(s)
- Khalid Al Sulaiman
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia.
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
- King Abdulaziz Medical City (KAMC)-Ministry of National Guard Health Affairs (MNGHA), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, PO Box 22490, Riyadh, 11426, Saudi Arabia.
| | - Ohoud Aljuhani
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalid Bin Saleh
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdulaziz Medical City (KAMC)-Ministry of National Guard Health Affairs (MNGHA), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, PO Box 22490, Riyadh, 11426, Saudi Arabia
| | - Hisham A Badreldin
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdulaziz Medical City (KAMC)-Ministry of National Guard Health Affairs (MNGHA), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, PO Box 22490, Riyadh, 11426, Saudi Arabia
| | - Abdullah Al Harthi
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Abdulaziz Medical City (KAMC)-Ministry of National Guard Health Affairs (MNGHA), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, PO Box 22490, Riyadh, 11426, Saudi Arabia
| | - Mohammed Alenazi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Aisha Alharbi
- Pharmaceutical Care Department, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Rahmah Algarni
- Pharmaceutical Care Department, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Shmeylan Al Harbi
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdulaziz Medical City (KAMC)-Ministry of National Guard Health Affairs (MNGHA), King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, PO Box 22490, Riyadh, 11426, Saudi Arabia
| | - Abdullah M Alhammad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ramesh Vishwakarma
- Biostatistics and Bioinformatics Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Sarah Aldekhyl
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Intensive Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- Biostatistics and Bioinformatics Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Exploring the Pharmacological Mechanisms of Tripterygium wilfordii Hook F against Cardiovascular Disease Using Network Pharmacology and Molecular Docking. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5575621. [PMID: 34435046 PMCID: PMC8382521 DOI: 10.1155/2021/5575621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/14/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022]
Abstract
Background Tripterygium wilfordii Hook F (TwHF) has been used in traditional Chinese medicine (TCM) for treating cardiovascular disease (CVD). However, the underlying pharmacological mechanisms of the effects of TwHF on CVD remain elusive. This study revealed the pharmacological mechanisms of TwHF acting on CVD based on a pharmacology approach. Materials and Methods The active compounds were selected from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database according to the absorption, distribution, metabolism, and excretion (ADME). The potential targets of TwHF were obtained from the SwissTargetPrediction database. The CVD-related therapeutic targets were collected from the DrugBank, the GeneCards database, and the OMIM database. Protein–protein interaction (PPI) network was generated by the STITCH database. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed by R package. The network of drug-targets-diseases-pathways was constructed by the Cytoscape software. Results The 41 effective ingredients of TwHF and the 178 common targets of TwHF and CVD-related were collected. Furthermore, AKT1, amyloid precursor protein (APP), mitogen-activated protein kinase 1 (MAPK), phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA), and cellular tumor antigen p53 (TP53) were identified as the core targets involved in the mechanism of TwHF on CVD. Top ten GO (biological processes, cellular components, and molecular functions) and KEGG pathways were screened with a P value ≤0.01. Finally, we constructed the network of TwHF-targets-CVD-GO-KEGG. Conclusions These findings demonstrate that the main active compound of TwHF, the core targets, and pathways maybe provide new insights into the development of a natural therapy for the prevention and treatment of CVD.
Collapse
|
36
|
Pan Q, Wu K, Tan J, Li Y, Liang X, Su M. Anti-neoplastic characteristics and potential targets of calycosin against bisphenol A-related osteosarcoma: bioinformatics analysis. Bioengineered 2021; 12:4278-4288. [PMID: 34311656 PMCID: PMC8806932 DOI: 10.1080/21655979.2021.1956401] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Environmentally, bisphenol A (BPA) is a well-known pollutant caused human health risk, including osteosarcoma (OS). OS, a deadly bone neoplasia, may occur in children and adults. However, the anti-OS pharmacotherapy prescribes limitedly in clinical practice. Interestingly, previous experimental evidences indicate calycosin-exerting potential anti-OS actions. Thus, in this report, we aimed to further characterize and detail the therapeutic targets and molecular mechanisms of calycosin-anti-BPA-related OS by using network pharmacology and molecular docking analyses. In results, the bioinformatics data disclosed all mapped, core targets, biological functions, molecular pathways of calycosin to treat BPA-related OS. The computational analysis using molecular docking indicated that potential binding ability of core targets in calycosin to treat BPA-related OS was identified. Moreover, detailed biological functions and optimal pathways of calycosin-anti-BPA-related OS were revealed, as shown in integrated network maps. Taken together, these network pharmacology and structural biology findings illustrate the core biotargets, pharmacological functions and pathways of calycosin-anti-BPA-related OS. Potentially, these core targets identified by molecular docking may attribute to the potential clinical application of calycosin against BPA-related OS.
Collapse
Affiliation(s)
- Qijin Pan
- Department of Oncology, Guigang City Peoples' Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, PR China
| | - Ka Wu
- Department of Pharmacy, The Second People's Hospital of Nanning City, the Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiachang Tan
- Department of Bone and Soft Tissue Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, PR China
| | - Yu Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Xiao Liang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Min Su
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| |
Collapse
|
37
|
Feng Z, Shi H, Liang B, Ge T, Cai M, Liu F, Huang K, Wen J, Chen Q, Ge B. Bioinformatics and experimental findings reveal the therapeutic actions and targets of pachymic acid against cystitis glandularis. Biofactors 2021; 47:665-673. [PMID: 33893687 DOI: 10.1002/biof.1734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/09/2021] [Indexed: 12/27/2022]
Abstract
Pachymic acid (PA), a bioactive ingredient isolated from Poria cocos Wolf, is reported with potential benefits of anti-inflammatory, anti-oxidative actions. It is reasoned that PA may play the potential benefits against cystitis glandularis (CG), an inflammation of the bladder tissue. In this study, we aimed to apply the network pharmacology and molecular docking analyses to reveal concrete anti-CG targets and mechanisms of PA, and then the bioinformatic findings were verified by using clinical and animal samples. The methodological data from network pharmacology approach showed that 303 and 243 reporting targets of CG and PA, and other 31 shared targets of CG and PA were identified. Subsequently, all top targets of PA against CG were screened out, including cyclooxygenase-2, epidermal growth factor receptor, tumor antigen p53 (TP53), tumor necrosis factor-alpha (TNF), interleukin-1 (IL-1) beta, proto-oncogene c-jun. Molecular docking data demonstrated that PA exerted potent bonding capacities with TNF, TP53 proteins in CG. In human study, the findings suggested that overactivated TNF-α expression and suppressed TP53 activation were detected in CG samples. In animal study, PA-treated mice showed reduced intravesical IL-1, IL-6 levels, and lactate dehydrogenase content, downregulated TNF-α and upregulated TP53 proteins in bladder samples. Taken together, our bioinformatics and experimental findings identify the key anti-CG biotargets and mechanisms of PA. More markedly, these pivotal pharmacological targets of PA against CG have been screened out and verified by using computational and experimental analyses.
Collapse
Affiliation(s)
- Zihao Feng
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Hailin Shi
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Bojian Liang
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Tianyu Ge
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Menghui Cai
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Feng Liu
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Kunping Huang
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jintao Wen
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Qiuhong Chen
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Bo Ge
- The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| |
Collapse
|
38
|
James PT, Ali Z, Armitage AE, Bonell A, Cerami C, Drakesmith H, Jobe M, Jones KS, Liew Z, Moore SE, Morales-Berstein F, Nabwera HM, Nadjm B, Pasricha SR, Scheelbeek P, Silver MJ, Teh MR, Prentice AM. The Role of Nutrition in COVID-19 Susceptibility and Severity of Disease: A Systematic Review. J Nutr 2021; 151:1854-1878. [PMID: 33982105 PMCID: PMC8194602 DOI: 10.1093/jn/nxab059] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/22/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Many nutrients have powerful immunomodulatory actions with the potential to alter susceptibility to coronavirus disease 2019 (COVID-19) infection, progression to symptoms, likelihood of severe disease, and survival. OBJECTIVE The aim was to review the latest evidence on how malnutrition across all its forms (under- and overnutrition and micronutrient status) may influence both susceptibility to, and progression of, COVID-19. METHODS We synthesized information on 13 nutrition-related components and their potential interactions with COVID-19: overweight, obesity, and diabetes; protein-energy malnutrition; anemia; vitamins A, C, D, and E; PUFAs; iron; selenium; zinc; antioxidants; and nutritional support. For each section we provide: 1) a landscape review of pertinent material; 2) a systematic search of the literature in PubMed and EMBASE databases, including a wide range of preprint servers; and 3) a screen of 6 clinical trial registries. All original research was considered, without restriction to study design, and included if it covered: 1) severe acute respiratory syndrome coronavirus (CoV) 2 (SARS-CoV-2), Middle East respiratory syndrome CoV (MERS-CoV), or SARS-CoV viruses and 2) disease susceptibility or 3) disease progression, and 4) the nutritional component of interest. Searches took place between 16 May and 11 August 2020. RESULTS Across the 13 searches, 2732 articles from PubMed and EMBASE, 4164 articles from the preprint servers, and 433 trials were returned. In the final narrative synthesis, we include 22 published articles, 38 preprint articles, and 79 trials. CONCLUSIONS Currently there is limited evidence that high-dose supplements of micronutrients will either prevent severe disease or speed up recovery. However, results of clinical trials are eagerly awaited. Given the known impacts of all forms of malnutrition on the immune system, public health strategies to reduce micronutrient deficiencies and undernutrition remain of critical importance. Furthermore, there is strong evidence that prevention of obesity and type 2 diabetes will reduce the risk of serious COVID-19 outcomes. This review is registered at PROSPERO as CRD42020186194.
Collapse
Affiliation(s)
- Philip T James
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Zakari Ali
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Bonell
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Carla Cerami
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Modou Jobe
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Kerry S Jones
- National Institute for Health Research (NIHR) Biomedical Research Centre (BRC) Nutritional Biomarker Laboratory, MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Zara Liew
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Sophie E Moore
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
- Department of Women and Children's Health, King's College London, London, United Kingdom
| | - Fernanda Morales-Berstein
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Helen M Nabwera
- Department of International Public Health, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Behzad Nadjm
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Pauline Scheelbeek
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Centre on Climate Change and Planetary Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Matt J Silver
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Megan R Teh
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew M Prentice
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
39
|
Liang X, Zhou R, Li Y, Yang L, Su M, Lai KP. Clinical characterization and therapeutic targets of vitamin A in patients with hepatocholangiocarcinoma and coronavirus disease. Aging (Albany NY) 2021; 13:15785-15800. [PMID: 34176789 PMCID: PMC8266307 DOI: 10.18632/aging.203220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
Recent reports indicate that patients with hepatocholangiocarcinoma (CHOL) have a higher morbidity and mortality rate for coronavirus disease (COVID-19). Anti-CHOL/COVID-19 medicines are inexistent. Vitamin A (VA) refers to a potent nutrient with anti-cytotoxic and anti-inflammatory actions. Therefore, this study aimed to determine the potential functions and molecular mechanisms of VA as a potential treatment for patients with both CHOL and COVID-19 (CHOL/COVID-19). The transcriptome data of CHOL patients were obtained from the Cancer Genome Analysis database. Furthermore, the network pharmacology approach and bioinformatics analysis were used to identify and reveal the molecular functions, therapeutic biotargets, and signaling of VA against CHOL/COVID-19. First, clinical findings identified the medical characteristics of CHOL patients with COVID-19, such as susceptibility gene, prognosis, recurrence, and survival rate. Anti-viral and anti-inflammatory pathways, and immunopotentiation were found as potential targets of VA against CHOL/COVID-19. These findings illustrated that VA may contribute to the clinical management of CHOL/COVID-19 achieved by induction of cell repair, suppression of oxidative stress and inflammatory reaction, and amelioration of immunity. Nine vital therapeutic targets (BRD2, NOS2, GPT, MAPK1, CXCR3, ICAM1, CDK4, CAT, and TMPRSS13) of VA against CHOL/COVID-19 were identified. For the first time, the potential pharmacological biotargets, function, and mechanism of action of VA in CHOL/COVID-19 were elucidated.
Collapse
Affiliation(s)
- Xiao Liang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, Guangxi, China.,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Rui Zhou
- Department of Hepatobiliary Surgery, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, China
| | - Yu Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, Guangxi, China.,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Lu Yang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, Guangxi, China.,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Min Su
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, Guangxi, China.,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, Guangxi, China.,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
40
|
Yu S, Wu K, Liang Y, Zhang H, Guo C, Yang B. Therapeutic targets and molecular mechanism of calycosin for the treatment of cerebral ischemia/reperfusion injury. Aging (Albany NY) 2021; 13:16804-16815. [PMID: 34176787 PMCID: PMC8266369 DOI: 10.18632/aging.203219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/14/2021] [Indexed: 12/22/2022]
Abstract
This study was designed to understand the pivotal anti-cerebral ischemia/reperfusion injury (CIRI) targets and pathways of calycosin through network pharmacology and molecular docking analyses. In this study, bioinformatics tools were employed to characterize and identify the pharmacological functions and mechanisms of calycosin for CIRI management. The network pharmacology data identified potential, merged CIRI-associated targets of calycosin including tumor protein p53 (TP53), protein kinase B (AKT1), vascular endothelial growth factor A (VEGFA), interleukin 6, tumor necrosis factor (TNF), and mitogen-activated protein kinase 1 (MAPK1). Molecular docking analysis indicated the binding efficacy of calycosin with three of the targets, namely TP53, AKT1, and VEGFA. The biological processes of calycosin for the treatment of CIRI are mainly involved in the improvement of endothelial cell proliferation and growth, inflammatory development, and cellular metabolism. In addition, the anti-CIRI actions of calycosin were primarily through suppression of the toll-like receptor, PI3K-AKT, TNF, MAPK, and VEGF signaling pathways. Taken together, the current bioinformatic findings revealed pivotal targets, biological functions, and pharmacological mechanisms of calycosin for the treatment of CIRI. In conclusion, calycosin, a functional phytoestrogen, can be potentially used for the treatment of CIRI in future clinical trials.
Collapse
Affiliation(s)
- Songzuo Yu
- Department of Neurosurgery, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, PR China
| | - Ka Wu
- Department of Pharmacy, The Second People's Hospital of Nanning City, The Third Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Yujia Liang
- College of Pharmacy, Guangxi Medical University, Nanning, PR China
| | - Haitao Zhang
- Department of Neurosurgery Area 1, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, PR China
| | - Chao Guo
- Department of Pharmacy, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, PR China
| | - Bin Yang
- College of Pharmacy, Guangxi Medical University, Nanning, PR China
| |
Collapse
|
41
|
Hong X, Li S, Wang J, Zhao Z, Feng Z. Circular RNA circFADS2 is overexpressed in sepsis and suppresses LPS-induced lung cell apoptosis by inhibiting the maturation of miR-15a-5p. BMC Immunol 2021; 22:29. [PMID: 33980140 PMCID: PMC8114495 DOI: 10.1186/s12865-021-00419-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Circular RNA circFADS2 plays protective roles in LPS-induced inflammation, which promotes sepsis, suggesting its involvement in sepsis. METHODS Expression of circFADS2, mature miR-15a-5p, and miR-15a-5p precursor in plasma samples from sepsis patients and healthy controls was determined by RT-qPCR. The circFADS2 expression vector was transfected in lung cells, followed by the measurement of the expression levels of mature miR-15a-5p and miR-15a-5p precursor to study the role of circFADS2 in miR-15a-5p maturation. Cell apoptosis was analyzed by cell apoptosis assay. RESULTS CircFADS2 was upregulated in sepsis and inversely correlated with mature miR-15a-5p, but not miR-15a-5p precursor. In lung cells, circFADS2 overexpression decreased the level of mature miR-15a-5p, but not miR-15a-5p precursor. LPS treatment decreased miR-15a-5p expression and increased circFADS2 level. Cell apoptosis analysis showed that circFADS2 overexpression reduced miR-15a-5p overexpression-induced apoptosis of LPS-treated lung cells. CONCLUSIONS CircFADS2 is upregulated in sepsis to suppress LPS-induced lung cell apoptosis by inhibiting miR-15a-5p maturation.
Collapse
Affiliation(s)
- Xiaoyang Hong
- Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, No. 5 Nanmencang, Dongshitiao, Dongcheng District, Beijing, 100700, P. R. China
| | - Shuanglei Li
- Department of Cardiovascular Surgery, PLA General Hospital, Beijing, 100853, P. R. China
| | - Jie Wang
- Surgical Pediatric Intensive Care Unit, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou City, Henan Province, 450018, P. R. China
| | - Zhe Zhao
- Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, No. 5 Nanmencang, Dongshitiao, Dongcheng District, Beijing, 100700, P. R. China
| | - Zhichun Feng
- Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, No. 5 Nanmencang, Dongshitiao, Dongcheng District, Beijing, 100700, P. R. China.
| |
Collapse
|
42
|
Li R, Li Y, Liang X, Yang L, Su M, Lai KP. Network Pharmacology and bioinformatics analyses identify intersection genes of niacin and COVID-19 as potential therapeutic targets. Brief Bioinform 2021; 22:1279-1290. [PMID: 33169132 PMCID: PMC7717147 DOI: 10.1093/bib/bbaa300] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Patients with colorectal cancer (CRC) may be susceptible to the coronavirus disease-2019 (COVID-19). However, anti-CRC/COVID-19 treatment options are currently unavailable. Since niacin is a vitamin with cytoprotective and anti-inflammatory functions, this study aimed to evaluate the possible functional roles and underlying mechanisms of action of niacin as an anti-COVID-19 and -CRC therapy. INTERVENTIONS We used a series of network pharmacology-based and computational analyses to understand and characterize the binding capacity, biological functions, pharmacological targets and therapeutic mechanisms of niacin in CRC/COVID-19. MEASUREMENTS AND MAIN RESULTS We revealed the clinical characteristics of CRC patients and COVID-19 patients, including predisposing genes, survival rate and prognosis. Moreover, the results of molecular docking analysis indicated that niacin exerted effective binding capacity in COVID-19. Further, we disclosed the targets, biological functions and signaling pathways of niacin in CRC/COVID-19. The analysis indicated that niacin could help in treating CRC/COVID-19 through cytoprotection, enhancement of immunologic functions, inhibition of inflammatory reactions and regulation of cellular microenvironment. Furthermore, five core pharmacological targets of niacin in CRC/COVID-19 were also identified, including BCL2L1, PTGS2, IL1B, IFNG and SERPINE1. CONCLUSIONS This study, for the first time, revealed the niacin-associated molecular functions and pharmacological targets for treating CRC/COVID-19, as COVID-19 remains a serious pandemic. But the findings were not validated in actual CRC patients infected with COVID-19, so further investigation is needed to confirm the potential use of niacin for treating CRC/COVID-19.
Collapse
Affiliation(s)
| | - Yu Li
- Gyuilin Medical University
| | | | | | - Min Su
- Gyuilin Medical University
| | | |
Collapse
|
43
|
Milani GP, Macchi M, Guz-Mark A. Vitamin C in the Treatment of COVID-19. Nutrients 2021; 13:nu13041172. [PMID: 33916257 PMCID: PMC8065688 DOI: 10.3390/nu13041172] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 01/29/2023] Open
Abstract
Vitamin C is an essential nutrient that serves as antioxidant and plays a major role as co-factor and modulator of various pathways of the immune system. Its therapeutic effect during infections has been a matter of debate, with conflicting results in studies of respiratory infections and in critically ill patients. This comprehensive review aimed to summarize the current evidence regarding the use of vitamin C in the prevention or treatment of patients with SARS-CoV2 infection, based on available publications between January 2020 and February 2021. Overall, 21 publications were included in this review, consisting of case-reports and case-series, observational studies, and some clinical trials. In many of the publications, data were incomplete, and in most clinical trials the results are still pending. No studies regarding prevention of COVID-19 with vitamin C supplementation were found. Although some clinical observations reported improved medical condition of patients with COVID-19 treated with vitamin C, available data from controlled studies are scarce and inconclusive. Based on the theoretical background presented in this article, and some preliminary encouraging studies, the role of vitamin C in the treatment of patients with SARS-CoV-2 infection should be further investigated.
Collapse
Affiliation(s)
- Gregorio Paolo Milani
- Pediatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.P.M.); (M.M.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Marina Macchi
- Pediatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.P.M.); (M.M.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Anat Guz-Mark
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Petach-Tikva 4920227, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-3-9253673
| |
Collapse
|
44
|
Lin Y, Luo L, Lin H, Li X, Huang R. Potential therapeutic targets and molecular details of anthocyan-treated inflammatory bowel disease: a systematic bioinformatics analysis of network pharmacology. RSC Adv 2021; 11:8239-8249. [PMID: 35423341 PMCID: PMC8695082 DOI: 10.1039/d0ra09117k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/03/2021] [Indexed: 12/20/2022] Open
Abstract
Anthocyans, containing anthocyanins and anthocyanidins, play a crucial role in preventing and treating inflammatory bowel disease (IBD). Most anthocyanins and their basic elements, namely anthocyanidins have been recognized for the effective treatment of IBD, but the key biomarkers of anthocyan-treated IBD remain unclear. In this study, a bioinformatics analysis based on network pharmacology was performed to demonstrate the core-targets, biological functions, and signaling pathways of most common anthocyanidins that existed in anthocyans to reveal their potential or major mechanisms. The network pharmacology of the multi-target drug molecular design with specific signal nodes was selected, which was used to analyse core targets and complete the bioinformatics analysis of core targets. The network assays indicated 44 common targeted genes, 5 of which were core targets of both six most common anthocyanidins and IBD. These 44 common targets related to major signaling mechanisms of the six most common anthocyanidins in IBD may involve following processes: promotion of intracellular metabolism and proliferation, inhibition of cell necrosis, anti-inflammation and regulation of intestinal epithelial survival mainly via pathways such as, the EGFR tyrosine kinase inhibitor resistance pathway, platelet activation, microRNAs in cancer, arachidonic acid metabolism and the cGMP-PKG signaling pathway. Thus, our findings may provide other molecular details about anthocyans in the treatment of IBD and contribute towards the use of anthocyanidins, which will be meaningful shedding light on the action mechanisms of anthocyanidins in treating IBD.
Collapse
Affiliation(s)
- Yuqi Lin
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University Guangzhou 510642 China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University Zhanjiang 524023 China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang Zhanjiang Guangdong 524023 China
| | - Haowen Lin
- The First Clinical College, Guangdong Medical University Zhanjiang 524023 China
| | - Xiaoling Li
- Animal Experiment Center, Guangdong Medical University Zhanjiang 524023 China
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University Guangzhou 510642 China
| |
Collapse
|
45
|
Zhu N, Huang B, Jiang W. Targets of Vitamin C With Therapeutic Potential for Cardiovascular Disease and Underlying Mechanisms: A Study of Network Pharmacology. Front Pharmacol 2021; 11:591337. [PMID: 33603661 PMCID: PMC7884818 DOI: 10.3389/fphar.2020.591337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Vitamin C (ascorbic acid) is a nutrient used to treat cardiovascular disease (CVD). However, the pharmacological targets of vitamin C and the mechanisms underlying the therapeutic effects of vitamin C on CVD remain to be elucidated. In this study, we used network pharmacology approach to investigate the pharmacological mechanisms of vitamin C for the treatment of CVD. The core targets, major hubs, enriched biological processes, and key signaling pathways were identified. A protein-protein interaction network and an interaction diagram of core target-related pathways were constructed. Three core targets were identified, including phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform, signal transducer and activator of transcription-3 (STAT3), and prothrombin. The GO and KEGG analyses identified top 20 enriched biological processes and signaling pathways involved in the therapeutic effects of vitamin C on CVD. The JAK-STAT, STAT, PD1, EGFR, FoxO, and chemokines signaling pathways may be highly involved in the protective effects of vitamin C against CVD. In conclusion, our bioinformatics analyses provided evidence on the possible therapeutic mechanisms of vitamin C in CVD treatment, which may contribute to the development of novel drugs for CVD.
Collapse
Affiliation(s)
- Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s Hospital, Wenzhou, China
| | - Bingwu Huang
- Department of Anesthesiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s Hospital, Wenzhou, China
| | - Wenbing Jiang
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s Hospital, Wenzhou, China
| |
Collapse
|
46
|
Yang L, Xu H, Chen Y, Miao C, Zhao Y, Xing Y, Zhang Q. Melatonin: Multi-Target Mechanism Against Diminished Ovarian Reserve Based on Network Pharmacology. Front Endocrinol (Lausanne) 2021; 12:630504. [PMID: 33959095 PMCID: PMC8095380 DOI: 10.3389/fendo.2021.630504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/29/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Diminished ovarian reserve (DOR) significantly increases the risk of female infertility and contributes to reproductive technology failure. Recently, the role of melatonin in improving ovarian reserve (OR) has attracted widespread attention. However, details on the pharmacological targets and mechanisms of melatonin-improved OR remain unclear. OBJECTIVE A systems pharmacology strategy was proposed to elucidate the potential therapeutic mechanism of melatonin on DOR at the molecular, pathway, and network levels. METHODS The systems pharmacological approach consisted of target identification, data integration, network construction, bioinformatics analysis, and molecular docking. RESULTS From the molecular perspective, 26 potential therapeutic targets were identified. They participate in biological processes related to DOR development, such as reproductive structure development, epithelial cell proliferation, extrinsic apoptotic signaling pathway, PI3K signaling, among others. Eight hub targets (MAPK1, AKT1, EGFR, HRAS, SRC, ESR1, AR, and ALB) were identified. From the pathway level, 17 significant pathways, including the PI3K-Akt signaling pathway and the estrogen signaling pathway, were identified. In addition, the 17 signaling pathways interacted with the 26 potential therapeutic targets to form 4 functional modules. From the network point of view, by regulating five target subnetworks (aging, cell growth and death, development and regeneration, endocrine and immune systems), melatonin could exhibit anti-aging, anti-apoptosis, endocrine, and immune system regulation effects. The molecular docking results showed that melatonin bound well to all hub targets. CONCLUSION This study systematically and intuitively illustrated the possible pharmacological mechanisms of OR improvement by melatonin through anti-aging, anti-apoptosis, endocrine, and immune system regulation effects.
Collapse
Affiliation(s)
- Liuqing Yang
- Department of Traditional Chinese Medical Gynecology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongbin Xu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Chen
- Department of Traditional Chinese Medical Gynecology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenyun Miao
- Department of Traditional Chinese Medical Gynecology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhao
- Department of Traditional Chinese Medical Gynecology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Xing
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qin Zhang
- Department of Traditional Chinese Medical Gynecology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Qin Zhang,
| |
Collapse
|
47
|
Li Y, Yu S, Li Y, Liang X, Su M, Li R. Medical Significance of Uterine Corpus Endometrial Carcinoma Patients Infected With SARS-CoV-2 and Pharmacological Characteristics of Plumbagin. Front Endocrinol (Lausanne) 2021; 12:714909. [PMID: 34712201 PMCID: PMC8547653 DOI: 10.3389/fendo.2021.714909] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/08/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Clinically, evidence shows that uterine corpus endometrial carcinoma (UCEC) patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may have a higher death-rate. However, current anti-UCEC/coronavirus disease 2019 (COVID-19) treatment is lacking. Plumbagin (PLB), a pharmacologically active alkaloid, is an emerging anti-cancer inhibitor. Accordingly, the current report was designed to identify and characterize the anti-UCEC function and mechanism of PLB in the treatment of patients infected with SARS-CoV-2 via integrated in silico analysis. METHODS The clinical analyses of UCEC and COVID-19 in patients were conducted using online-accessible tools. Meanwhile, in silico methods including network pharmacology and biological molecular docking aimed to screen and characterize the anti-UCEC/COVID-19 functions, bio targets, and mechanisms of the action of PLB. RESULTS The bioinformatics data uncovered the clinical characteristics of UCEC patients infected with SARS-CoV-2, including specific genes, health risk, survival rate, and prognostic index. Network pharmacology findings disclosed that PLB-exerted anti-UCEC/COVID-19 effects were achieved through anti-proliferation, inducing cytotoxicity and apoptosis, anti-inflammation, immunomodulation, and modulation of some of the key molecular pathways associated with anti-inflammatory and immunomodulating actions. Following molecular docking analysis, in silico investigation helped identify the anti-UCEC/COVID-19 pharmacological bio targets of PLB, including mitogen-activated protein kinase 3 (MAPK3), tumor necrosis factor (TNF), and urokinase-type plasminogen activator (PLAU). CONCLUSIONS Based on the present bioinformatic and in silico findings, the clinical characterization of UCEC/COVID-19 patients was revealed. The candidate, core bio targets, and molecular pathways of PLB action in the potential treatment of UCEC/COVID-19 were identified accordingly.
Collapse
Affiliation(s)
- Yongming Li
- Department of Gynecology, Guigang Maternal and Child Health Care Hospital, Guigang, China
| | - Songzuo Yu
- Department of Neurosurgery, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Yu Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Xiao Liang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Min Su
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
- *Correspondence: Min Su, ; Rong Li, ;
| | - Rong Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
- *Correspondence: Min Su, ; Rong Li, ;
| |
Collapse
|
48
|
Ye Y, Huang Z, Chen M, Mo Y, Mo Z. Luteolin Potentially Treating Prostate Cancer and COVID-19 Analyzed by the Bioinformatics Approach: Clinical Findings and Drug Targets. Front Endocrinol (Lausanne) 2021; 12:802447. [PMID: 35178029 PMCID: PMC8844187 DOI: 10.3389/fendo.2021.802447] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a serious epidemic, characterized by potential mutation and can bring about poor vaccine efficiency. It is evidenced that patients with malignancies, including prostate cancer (PC), may be highly vulnerable to the SARS-CoV-2 infection. Currently, there are no existing drugs that can cure PC and COVID-19. Luteolin can potentially be employed for COVID-19 treatment and serve as a potent anticancer agent. Our present study was conducted to discover the possible drug target and curative mechanism of luteolin to serve as treatment for PC and COVID-19. The differential gene expression of PC cases was determined via RNA sequencing. The application of network pharmacology and molecular docking aimed to exhibit the drug targets and pharmacological mechanisms of luteolin. In this study, we found the top 20 up- and downregulated gene expressions in PC patients. Enrichment data demonstrated anti-inflammatory effects, where improvement of metabolism and enhancement of immunity were the main functions and mechanism of luteolin in treating PC and COVID-19, characterized by associated signaling pathways. Additional core drug targets, including MPO and FOS genes, were computationally identified accordingly. In conclusion, luteolin may be a promising treatment for PC and COVID-19 based on bioinformatics findings, prior to future clinical validation and application.
Collapse
Affiliation(s)
- Yu Ye
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziyan Huang
- Health Management Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Manying Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongfeng Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Zengnan Mo,
| |
Collapse
|
49
|
Qin X, Huang C, Wu K, Li Y, Liang X, Su M, Li R. Anti-coronavirus disease 2019 (COVID-19) targets and mechanisms of puerarin. J Cell Mol Med 2020; 25:677-685. [PMID: 33241658 PMCID: PMC7753316 DOI: 10.1111/jcmm.16117] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/29/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The present study aimed to uncover the pharmacological function and underlying mechanism of puerarin as a potential treatment for COVID‐19, using an in silico methodology, including network pharmacology and molecular docking. The pivotal targets of puerarin to treat COVID‐19 were identified and included the epidermal growth factor receptor (EGFR), tumour necrosis factor (TNF), tumour protein p53 (TP53), caspase 3 (CASP3), RELA proto‐oncogene (RELA), Fos proto‐oncogene (FOS), caspase 8 (CASP8), prostaglandin‐endoperoxide synthase 2 (PTGS2), interleukin 2 (IL2), protein kinase CB (PRKCB), B cell lymphoma/leukaemia gene‐2 (BCL2), protein kinase CA (PRKCA), nitric oxide synthase 3 (NOS3) and peroxisome proliferator–activated receptor gamma (PPARG). Functionally, the anti–COVID‐19 action of puerarin was associated with the suppression of oxidative stress and inflammatory cascades, and cell apoptosis. The signalling pathways of puerarin to treat COVID‐19 included modulation of the pathways of apoptosis, IL‐17 signalling, mitogen‐activated protein kinase (MAPK) signalling and TNF signalling. Molecular docking data illustrated the binding capacity of puerarin with COVID‐19 and the effective anti–COVID‐19 activity of puerarin. Taken together, our current network pharmacology–based findings revealed the pharmacological role of puerarin in the treatment of COVID‐19. Furthermore, the bioinformatic findings elucidated that some of these pivotal targets might serve as potential molecular markers for detecting COVID‐19.
Collapse
Affiliation(s)
- Xingyue Qin
- Department of Neurology (Area Two), Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Chen Huang
- The Center for Data Science in Health and Medicine, Business School, Qingdao University, Qingdao, China
| | - Ka Wu
- Department of Pharmacy, The Second People's Hospital of Nanning City, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Li
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Xiao Liang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Min Su
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Rong Li
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| |
Collapse
|
50
|
Abobaker A, Alzwi A, Alraied AHA. Overview of the possible role of vitamin C in management of COVID-19. Pharmacol Rep 2020; 72:1517-1528. [PMID: 33113146 PMCID: PMC7592143 DOI: 10.1007/s43440-020-00176-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
The mainstay of management of coronavirus disease 2019 (COVID-19) is mainly supportive as to date there is no effective antiviral treatment, apart from remdesivir which has been approved by Food and Drug administration (FDA) for treatment of COVID-19, or vaccine. Supplementation with micronutrients, such as vitamins and minerals, has gained an increasing interest as part of the supportive management of COVID-19. Vitamin C levels in serum and leukocytes are depleted during the acute stage of infection owing to increased metabolic demands. High-dose vitamin C supplement helps to normalise both serum and leukocytes vitamin C levels. Vitamin C has multiple pharmacological characteristics, antiviral, anti-oxidant, anti-inflammatory and immunomodulatory effects, which make it a potential therapeutic option in management of COVID-19. The use of high dose of intravenous vitamin C for management of COVID-19 in China and the United Stated has shown promising results. There were no reported adverse reactions with the short-term use of high dose of vitamin C. Given the fact that vitamin C is cheap, available and safe drug with beneficial effects in management of viral infections and critically ill patients reported in previous clinical trials, it is sensible to add it to COVID-19 management protocol particularly if the current ongoing clinical trials testing the effect of vitamin C in management of COVID-19 show positive results.
Collapse
Affiliation(s)
- Anis Abobaker
- Spire Fylde Coast Hospital, St Walburgas road, Blackpool, FY3 8BP, UK.
| | | | | |
Collapse
|