1
|
Ma J, Wei P, Xu X, Dong R, Deng X, Zhang F, Sun M, Li M, Liu W, Yao J, Cao Y, Ying L, Yang Y, Yang Y, Wu X, She G. Machine learning-assisted analysis of serum metabolomics and network pharmacology reveals the effective compound from herbal formula against alcoholic liver injury. Chin Med 2025; 20:48. [PMID: 40217538 PMCID: PMC11992827 DOI: 10.1186/s13020-025-01094-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The popularity of herbal formulas is increasing worldwide. Nevertheless, the effective compound is challenging to identify due to its intricate composition and multiple targets. METHODS An integration machine learning-assisted approach was established, whereby the particular action mechanism and direct target were obtained through the correlation of compounds, targets, and metabolites. The association between a compound and an action pathway was selected from the shortest path of the "compound-target-pathway-disease" network, which was analyzed using the Floyd-Warshall algorithm. Subsequently, an investigation was conducted into the relationship between metabolites and action pathways, as well as targets, through the analysis of serum metabolomic profiling and the selection of metabolite biomarkers by random forest. In order to accurately identify the direct acting target as well as the most effective compound, the relationship between the compounds and their targets was investigated using a feature-based prediction model conducted by AdaBoost. The binding mode of the effective compound and the direct-acting target was verified by molecular docking, dynamics simulations, and western blotting. In this study, Baiji Wuweizi Granule (BWG) was employed to elucidate the effective compound against alcoholic liver injury (ALD). RESULTS BWG exerted an influence on the serum metabolomic, resulting in the identification of seven potential biomarkers. Furthermore, six effective compounds and the PI3K-AKT signalling pathway were identified through a co-analysis with the shortest path from compound to ALD in the "compound-target-pathway-disease" network. It was postulated that the effective compounds would bind with key targets from the PI3K-AKT signaling pathway, as indicated by the prediction model of compound-target interaction (R2 > 0.95). The dominant bonding type for the effective compounds and key targets was hydrogen bond. These results indicated that AKT1 was the notable target for BWG, and that 2,3,4,7-tetramethoxyphenanthrene was the marker compound for BWG against ALD. The present study provides evidence that the protective effect of BWG on ALD can be mediated by the PI3K-AKT signaling pathway. CONCLUSIONS Our findings demonstrate the value of a machine learning-assisted approach in identifying the key compound, target and pathway that underpin the efficacy of an herbal formula. This provides a foundation for future clinical and fundamental research.
Collapse
Affiliation(s)
- Jiamu Ma
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Peng Wei
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Xiao Xu
- Analysis and Test Center, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Ruijuan Dong
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Xixi Deng
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Feng Zhang
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Mengyu Sun
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Mingxia Li
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Wei Liu
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Jianling Yao
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Yu Cao
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Letian Ying
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Yuqing Yang
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Yongqi Yang
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China
| | - Xiaopeng Wu
- Analysis and Test Center, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China.
| | - Gaimei She
- Beijing University of Chinese Medicine, Fangshan District, Beijing, 100029, China.
| |
Collapse
|
2
|
Li Y, Shen Y, Cai Y, Zhang Y, Gao J, Huang L, Si W, Zhou K, Gao S, Luo Q. Integrating transcriptomic data with a novel drug efficacy prediction model for TCM active compound discovery. Sci Rep 2025; 15:7688. [PMID: 40044718 PMCID: PMC11882833 DOI: 10.1038/s41598-024-82498-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/03/2024] [Indexed: 03/09/2025] Open
Abstract
Identifying the active natural compounds remains a challenge for drug discovery, and new algorithms need to be developed to predict active ingredients from complex natural products. Here, we proposed Meta-DEP, a Meta-paths-based Drug Efficacy Prediction based on drug-protein-disease heterogeneity network, where Meta-paths contain all the shortest paths between drug targets and disease-related proteins in the network and drug efficacy is measured by a predictive score according to drug disease network proximity. Experiments show that Meta-DEP performs better than traditional network topology analysis on drug-disease interaction prediction task. Further investigations demonstrate that the key targets identified by Meta-DEP for drug efficacy are consistent with clinical pharmacological evidence. To prove that Meta-DEP can be used to discover active natural compounds, we apply it to predict the relationship between the monomeric components of traditional Chinese medicine included in the TCMSP database and diseases. Results indicate that Meta-DEP can accurately predict most of the drug-disease pairs included in the TCMSP database. In addition, biological experiments are directly used to demonstrate that Meta-DEP can mined active compound from traditional Chinese medicine with integrating disease transcriptomic data. Overall, the model developed in this study provides new impetus for driving the natural compound into innovative lead molecule. Code and data are available at https://github.com/t9lex/Meta-DEP .
Collapse
Affiliation(s)
- Yingcan Li
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
- Research Center for Neurological Disorders, School of Basic Medicine, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yu Shen
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Yezi Cai
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
- Research Center for Neurological Disorders, School of Basic Medicine, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yulin Zhang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Jiahui Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
- Research Center for Neurological Disorders, School of Basic Medicine, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Lei Huang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Weinuo Si
- Research Center for Neurological Disorders, School of Basic Medicine, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Kai Zhou
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China.
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China.
| | - Qichao Luo
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China.
- Research Center for Neurological Disorders, School of Basic Medicine, Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
3
|
Tie D, He M, Li W, Xiang Z. Advances in the application of network analysis methods in traditional Chinese medicine research. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156256. [PMID: 39615211 DOI: 10.1016/j.phymed.2024.156256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/03/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVE This review aims at evaluating the role and potential applications of network analysis methods in the medicinal substances of traditional Chinese medicine (TCM), theories of TCM compatibility, properties of herbs, and TCM syndromes. METHODS Literature was retrieved from databases, such as CNKI, PubMed, and Web of Science, using keywords, including "network analysis," "network biology," "network pharmacology," and "network medicine." The extracted literature included the biological network construction (including ingredient-target and target-disease relations), analysis of network topology characteristics (including node degree, clustering coefficient, and path length), network modularization analysis, functional annotation and so on. These studies were categorized and organized based on their research methods, application domains, and other relevant characteristics. RESULTS Network analysis algorithms, such as network distance, random walk, matrix factorization, graph embedding, and graph neural networks, are widely applied in fields related to the properties, compatibility, and mechanisms of TCM. They effectively reflect the interactive relations within the complex systems of TCM and elucidate and clarify theories, such as the effective substances, the principles of TCM compatibility, the TCM syndromes, and the properties of TCM. CONCLUSION The network analysis method is a powerful mathematical and computational tool that reveals the structure, dynamics, and functions of complex systems by analyzing the elements and their relations. This approach has effectively promoted the modernization of TCM, providing essential theoretical and practical tools for personalized treatment and scientific research on TCM. It also offers a significant methodological framework for the modernization and internationalization of TCM.
Collapse
Affiliation(s)
- Defu Tie
- Medical School, Hangzhou City University, Hangzhou, 310015, China; College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Mulan He
- Medical School, Hangzhou City University, Hangzhou, 310015, China; College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Wenlong Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Zheng Xiang
- Medical School, Hangzhou City University, Hangzhou, 310015, China.
| |
Collapse
|
4
|
Brattig-Correia R, Almeida JM, Wyrwoll MJ, Julca I, Sobral D, Misra CS, Di Persio S, Guilgur LG, Schuppe HC, Silva N, Prudêncio P, Nóvoa A, Leocádio AS, Bom J, Laurentino S, Mallo M, Kliesch S, Mutwil M, Rocha LM, Tüttelmann F, Becker JD, Navarro-Costa P. The conserved genetic program of male germ cells uncovers ancient regulators of human spermatogenesis. eLife 2024; 13:RP95774. [PMID: 39388236 PMCID: PMC11466473 DOI: 10.7554/elife.95774] [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] [Indexed: 10/12/2024] Open
Abstract
Male germ cells share a common origin across animal species, therefore they likely retain a conserved genetic program that defines their cellular identity. However, the unique evolutionary dynamics of male germ cells coupled with their widespread leaky transcription pose significant obstacles to the identification of the core spermatogenic program. Through network analysis of the spermatocyte transcriptome of vertebrate and invertebrate species, we describe the conserved evolutionary origin of metazoan male germ cells at the molecular level. We estimate the average functional requirement of a metazoan male germ cell to correspond to the expression of approximately 10,000 protein-coding genes, a third of which defines a genetic scaffold of deeply conserved genes that has been retained throughout evolution. Such scaffold contains a set of 79 functional associations between 104 gene expression regulators that represent a core component of the conserved genetic program of metazoan spermatogenesis. By genetically interfering with the acquisition and maintenance of male germ cell identity, we uncover 161 previously unknown spermatogenesis genes and three new potential genetic causes of human infertility. These findings emphasize the importance of evolutionary history on human reproductive disease and establish a cross-species analytical pipeline that can be repurposed to other cell types and pathologies.
Collapse
Affiliation(s)
- Rion Brattig-Correia
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Department of Systems Science and Industrial Engineering, Binghamton UniversityNew YorkUnited States
| | - Joana M Almeida
- Instituto Gulbenkian de CiênciaOeirasPortugal
- EvoReproMed Lab, Environmental Health Institute (ISAMB), Associate Laboratory TERRA, Faculty of Medicine, University of LisbonLisbonPortugal
| | - Margot Julia Wyrwoll
- Centre of Medical Genetics, Institute of Reproductive Genetics, University and University Hospital of MünsterMünsterGermany
| | - Irene Julca
- School of Biological Sciences, Nanyang Technological UniversitySingaporeSingapore
| | - Daniel Sobral
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University LisbonLisbonPortugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University LisbonCaparicaPortugal
| | - Chandra Shekhar Misra
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de LisboaOeirasPortugal
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital MünsterMünsterGermany
| | | | - Hans-Christian Schuppe
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig-UniversityGiessenGermany
| | - Neide Silva
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Pedro Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de LisboaLisboaPortugal
| | - Ana Nóvoa
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | | | - Joana Bom
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Sandra Laurentino
- Centre of Reproductive Medicine and Andrology, University Hospital MünsterMünsterGermany
| | | | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital MünsterMünsterGermany
| | - Marek Mutwil
- School of Biological Sciences, Nanyang Technological UniversitySingaporeSingapore
| | - Luis M Rocha
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Department of Systems Science and Industrial Engineering, Binghamton UniversityNew YorkUnited States
| | - Frank Tüttelmann
- Centre of Medical Genetics, Institute of Reproductive Genetics, University and University Hospital of MünsterMünsterGermany
| | - Jörg D Becker
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de LisboaOeirasPortugal
| | - Paulo Navarro-Costa
- Instituto Gulbenkian de CiênciaOeirasPortugal
- EvoReproMed Lab, Environmental Health Institute (ISAMB), Associate Laboratory TERRA, Faculty of Medicine, University of LisbonLisbonPortugal
| |
Collapse
|
5
|
Liao X, Ozcan M, Shi M, Kim W, Jin H, Li X, Turkez H, Achour A, Uhlén M, Mardinoglu A, Zhang C. Open MoA: revealing the mechanism of action (MoA) based on network topology and hierarchy. Bioinformatics 2023; 39:btad666. [PMID: 37930015 PMCID: PMC10637856 DOI: 10.1093/bioinformatics/btad666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
MOTIVATION Many approaches in systems biology have been applied in drug repositioning due to the increased availability of the omics data and computational biology tools. Using a multi-omics integrated network, which contains information of various biological interactions, could offer a more comprehensive inspective and interpretation for the drug mechanism of action (MoA). RESULTS We developed a computational pipeline for dissecting the hidden MoAs of drugs (Open MoA). Our pipeline computes confidence scores to edges that represent connections between genes/proteins in the integrated network. The interactions showing the highest confidence score could indicate potential drug targets and infer the underlying molecular MoAs. Open MoA was also validated by testing some well-established targets. Additionally, we applied Open MoA to reveal the MoA of a repositioned drug (JNK-IN-5A) that modulates the PKLR expression in HepG2 cells and found STAT1 is the key transcription factor. Overall, Open MoA represents a first-generation tool that could be utilized for predicting the potential MoA of repurposed drugs and dissecting de novo targets for developing effective treatments. AVAILABILITY AND IMPLEMENTATION Source code is available at https://github.com/XinmengLiao/Open_MoA.
Collapse
Affiliation(s)
- Xinmeng Liao
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
| | - Mehmet Ozcan
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
- Department of Medical Biochemistry, Faculty of Medicine, Zonguldak Bulent Ecevit University, 67630 Zonguldak, Turkey
| | - Mengnan Shi
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
| | - Woonghee Kim
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
| | - Han Jin
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
| | - Xiangyu Li
- Guangzhou National Laboratory, Guangzhou, Guangdong Province 510005, China
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine, Solna, Karolinska Institute, 17176 Stockholm, Sweden
| | - Mathias Uhlén
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
| | - Adil Mardinoglu
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, United Kingdom
| | - Cheng Zhang
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, 17121 Stockholm, Sweden
| |
Collapse
|
6
|
Identifying Tumor-Associated Genes from Bilayer Networks of DNA Methylation Sites and RNAs. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010076. [PMID: 36676027 PMCID: PMC9861397 DOI: 10.3390/life13010076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022]
Abstract
Network theory has attracted much attention from the biological community because of its high efficacy in identifying tumor-associated genes. However, most researchers have focused on single networks of single omics, which have less predictive power. With the available multiomics data, multilayer networks can now be used in molecular research. In this study, we achieved this with the construction of a bilayer network of DNA methylation sites and RNAs. We applied the network model to five types of tumor data to identify key genes associated with tumors. Compared with the single network, the proposed bilayer network resulted in more tumor-associated DNA methylation sites and genes, which we verified with prognostic and KEGG enrichment analyses.
Collapse
|
7
|
Yang J, Li Z, Wu WKK, Yu S, Xu Z, Chu Q, Zhang Q. Deep learning identifies explainable reasoning paths of mechanism of action for drug repurposing from multilayer biological network. Brief Bioinform 2022; 23:6809964. [PMID: 36347526 DOI: 10.1093/bib/bbac469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/07/2022] [Accepted: 09/29/2022] [Indexed: 11/11/2022] Open
Abstract
The discovery and repurposing of drugs require a deep understanding of the mechanism of drug action (MODA). Existing computational methods mainly model MODA with the protein-protein interaction (PPI) network. However, the molecular interactions of drugs in the human body are far beyond PPIs. Additionally, the lack of interpretability of these models hinders their practicability. We propose an interpretable deep learning-based path-reasoning framework (iDPath) for drug discovery and repurposing by capturing MODA on by far the most comprehensive multilayer biological network consisting of the complex high-dimensional molecular interactions between genes, proteins and chemicals. Experiments show that iDPath outperforms state-of-the-art machine learning methods on a general drug repurposing task. Further investigations demonstrate that iDPath can identify explicit critical paths that are consistent with clinical evidence. To demonstrate the practical value of iDPath, we apply it to the identification of potential drugs for treating prostate cancer and hypertension. Results show that iDPath can discover new FDA-approved drugs. This research provides a novel interpretable artificial intelligence perspective on drug discovery.
Collapse
Affiliation(s)
- Jiannan Yang
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shi Yu
- The USC Norris Center for Cancer Drug Development, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhongzhi Xu
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| | - Qian Chu
- Department of Thoracic Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
8
|
Zheng YX, Wang KX, Chen SJ, Liao MX, Chen YP, Guan DG, Wu J, Xiong K. Decoding the Key Functional Combined Components Group and Uncovering the Molecular Mechanism of Longdan Xiegan Decoction in Treating Uveitis. Drug Des Devel Ther 2022; 16:3991-4011. [PMID: 36420429 PMCID: PMC9677932 DOI: 10.2147/dddt.s385136] [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: 08/23/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Longdan Xiegan Decoction (LXD) is a famous herbal formula in China. It has been proved that LXD has been shown to have a significant inhibitory effect on suppresses the inflammatory cells associated with uveitis. However, the key functional combination of component groups and their possible mechanisms remain unclear. Methods The community detecting model of the network, the functional response space, and reverse prediction model were utilized to decode the key components group (KCG) and possible mechanism of LXD in treating uveitis. Finally, MTT assay, NO assay and ELISA assay were applied to verify the effectiveness of KCG and the accuracy of our strategy. Results In the components-targets-pathogenic genes-disease (CTP) network, a combination of Huffman coding and random walk algorithm was used and eight foundational acting communities (FACs) were discovered with important functional significance. Verification has shown that FACs can represent the corresponding C-T network for treating uveitis. A novel node importance calculation method was designed to construct the functional response space and pick out 349 effective proteins. A total of 54 components were screened and defined as KCG. The pathway enrichment results showed that KCG and their targets enriched signal pathways of IL-17, Toll-like receptor, and T cell receptor played an important role in the pathogenesis of uveitis. Furthermore, experimental verification results showed that important KCG quercetin and sitosterol markedly inhibited the production of nitric oxide and significantly regulated the level of TNF-α and IFN-γ in Lipopolysaccharide-induced RAW264.7 cells. Discussion In this research, we decoded the potential mechanism of the multi-components-genes-pathways of LXD’s pharmacological action mode against uveitis based on an integrated pharmacology approach. The results provided a new perspective for the future studies of the anti-uveitis mechanism of traditional Chinese medicine.
Collapse
Affiliation(s)
- Yi-Xu Zheng
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Ke-Xin Wang
- Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, People’s Republic of China
| | - Si-Jin Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Mu-Xi Liao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, People’s Republic of China
| | - Yu-Peng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, People’s Republic of China
| | - Dao-Gang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jing Wu
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Correspondence: Jing Wu; Ke Xiong, Email ;
| | - Ke Xiong
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
9
|
Omontese BO, Sharma AK, Davison S, Jacobson E, DiConstanzo A, Webb MJ, Gomez A. Microbiome network traits in the rumen predict average daily gain in beef cattle under different backgrounding systems. Anim Microbiome 2022; 4:25. [PMID: 35346381 PMCID: PMC8961956 DOI: 10.1186/s42523-022-00175-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/20/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Backgrounding (BKG), the stage between weaning and finishing, significantly impacts feedlot performance in beef cattle; however, the contributions of the rumen microbiome to this growth stage remain unexplored. A longitudinal study was designed to assess how BKG affects rumen bacterial communities and average daily gain (ADG) in beef cattle. At weaning, 38 calves were randomly assigned to three BKG systems for 55 days (d): a high roughage diet within a dry lot (DL, n = 13); annual cover crop within a strip plot (CC, n = 13); and perennial pasture vegetation within rotational paddocks (PP, n = 12), as before weaning. After BKG, all calves were placed in a feedlot for 142 d and finished with a high energy ration. Calves were weighed periodically from weaning to finishing to determine ADG. Rumen bacterial communities were profiled by collecting fluid samples via oral probe and sequencing the V4 region of the 16S rRNA bacterial gene, at weaning, during BKG and finishing. RESULTS Rumen bacterial communities diverged drastically among calves once they were placed in each BKG system, including sharp decreases in alpha diversity for CC and DL calves only (P < 0.001). During BKG, DL calves showed a substantial increase of Proteobacteria (Succinivibrionaceae family) (P < 0.001), which also corresponded with greater ADG (P < 0.05). At the finishing stage, Proteobacteria bloomed for all calves, with no previous alpha or beta diversity differences being retained between groups. However, at finishing, PP calves showed a compensatory ADG, particularly greater than that in calves coming from DL BKG (P = 0.02). Microbiome network traits such as lower average shortest path length, and increased neighbor connectivity, degree, number and strength of bacterial interactions between rumen bacteria better predicted ADG during BKG and finishing than variation in specific taxonomic profiles. CONCLUSIONS Bacterial co-abundance interactions, as measured by network theory approaches, better predicted growth performance in beef cattle during BKG and finishing, than the abundance of specific taxa. These findings underscore the importance of early post weaning stages as potential targets for feeding interventions that can enhance metabolic interactions between rumen bacteria, to increase productive performance in beef cattle.
Collapse
Affiliation(s)
- Bobwealth O Omontese
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA
- Department of Food and Animal Sciences, Alabama A&M University, Normal, AL, 35762, USA
| | - Ashok K Sharma
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA
| | - Samuel Davison
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA
| | - Emily Jacobson
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA
| | - Alfredo DiConstanzo
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA
| | - Megan J Webb
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA
- Community Engagement and Partnerships, Eastern West Virginia Community and Technical College, Moorefield, WV, 26836, USA
| | - Andres Gomez
- Department of Animal Science, University of Minnesota, Saint Paul, MN, 55108, USA.
| |
Collapse
|
10
|
Pavel A, del Giudice G, Federico A, Di Lieto A, Kinaret PAS, Serra A, Greco D. Integrated network analysis reveals new genes suggesting COVID-19 chronic effects and treatment. Brief Bioinform 2021; 22:1430-1441. [PMID: 33569598 PMCID: PMC7929418 DOI: 10.1093/bib/bbaa417] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/13/2020] [Accepted: 12/19/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 disease led to an unprecedented health emergency, still ongoing worldwide. Given the lack of a vaccine or a clear therapeutic strategy to counteract the infection as well as its secondary effects, there is currently a pressing need to generate new insights into the SARS-CoV-2 induced host response. Biomedical data can help to investigate new aspects of the COVID-19 pathogenesis, but source heterogeneity represents a major drawback and limitation. In this work, we applied data integration methods to develop a Unified Knowledge Space (UKS) and used it to identify a new set of genes associated with SARS-CoV-2 host response, both in vitro and in vivo. Functional analysis of these genes reveals possible long-term systemic effects of the infection, such as vascular remodelling and fibrosis. Finally, we identified a set of potentially relevant drugs targeting proteins involved in multiple steps of the host response to the virus.
Collapse
Affiliation(s)
- Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Giusy del Giudice
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Antonio Di Lieto
- Department of Forensic Psychiatry, Aarhus University, Aarhus, Denmark
| | - Pia A S Kinaret
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Liu C, Ma Y, Zhao J, Nussinov R, Zhang YC, Cheng F, Zhang ZK. Computational network biology: Data, models, and applications. PHYSICS REPORTS 2020; 846:1-66. [DOI: 10.1016/j.physrep.2019.12.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|