351
|
Branco I, Choupina A. Bioinformatics: new tools and applications in life science and personalized medicine. Appl Microbiol Biotechnol 2021; 105:937-951. [PMID: 33404829 DOI: 10.1007/s00253-020-11056-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 11/29/2020] [Accepted: 12/09/2020] [Indexed: 11/28/2022]
Abstract
While we have a basic understanding of the functioning of the gene when coding sequences of specific proteins, we feel the lack of information on the role that DNA has on specific diseases or functions of thousands of proteins that are produced. Bioinformatics combines the methods used in the collection, storage, identification, analysis, and correlation of this huge and complex information. All this work produces an "ocean" of information that can only be "sailed" with the help of computerized methods. The goal is to provide scientists with the right means to explain normal biological processes, dysfunctions of these processes which give rise to disease and approaches that allow the discovery of new medical cures. Recently, sequencing platforms, a large scale of genomes and transcriptomes, have created new challenges not only to the genomics but especially for bioinformatics. The intent of this article is to compile a list of tools and information resources used by scientists to treat information from the massive sequencing of recent platforms to new generations and the applications of this information in different areas of life sciences including medicine. KEY POINTS: • Biological data mining • Omic approaches • From genotype to phenotype.
Collapse
Affiliation(s)
- Iuliia Branco
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253, Bragança, Portugal
| | - Altino Choupina
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253, Bragança, Portugal.
| |
Collapse
|
352
|
Jiao H, Yang H, Yan Z, Chen J, Xu M, Jiang Y, Liu Y, Xue Z, Ma Q, Li X, Chen J. Traditional Chinese Formula Xiaoyaosan Alleviates Depressive-Like Behavior in CUMS Mice by Regulating PEBP1-GPX4-Mediated Ferroptosis in the Hippocampus. Neuropsychiatr Dis Treat 2021; 17:1001-1019. [PMID: 33854318 PMCID: PMC8039849 DOI: 10.2147/ndt.s302443] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND At present, the pathogenesis of depression is not fully understood, and nearly half of depression patients experience no obvious effects during treatment. This study aimed to establish a depression mouse model to explore the possible role of ferroptosis in the pathogenesis of depression, and observe the effects of Xiaoyaosan on PEBP1-GPX4-mediated ferroptosis in the hippocampus. METHODS Forty-eight male C57BL/6 mice were randomly divided into a control group, CUMS group, Xiaoyaosan group and fluoxetine group, and the model was established by chronic unpredictable mild stress (CUMS) for a successive 6 weeks. The medication procedure was performed from the 4th to the 6th week of modeling. The behavioral evaluations were measured to evaluate depressive-like behaviors. The expressions of GPX4, FTH1, ACSL4 and COX2 were detected as ferroptosis-related indicators. Then, the total iron and ferrous content in the hippocampus were measured. The levels of PEBP1 and ERK1/2 were observed, and the expressions of GFAP and IBA1 were also detected to measure the functions of astrocytes and microglia in the hippocampus. RESULTS Eight herbs of Xiaoyaosan had 133 active ingredients which could regulate the 43 ferroptosis-related genes in depression. After 6 weeks of modeling, the data showed that mice in the CUMS group had obvious depressive-like behaviors, and medication with Xiaoyaosan or fluoxetine could significantly improve the behavioral changes. The expressions of GPX4, FTH1, ACSL4, COX2, PEBP1, ERK1/2, GFAP and IBA1 changed in the CUMS group mice, while the total iron and ferrous content also changed. Xiaoyaosan and fluoxetine had obvious curative effects that could significantly alleviate the above changes in the hippocampus. CONCLUSION Our results revealed that the activation of ferroptosis might exist in the hippocampi of CUMS-induced mice. The PEBP1-GPX4-mediated ferroptosis could be involved in the antidepressant mechanism of Xiaoyaosan. It also implied that ferroptosis could become a new target for research into the depression mechanism and antidepressant drugs.
Collapse
Affiliation(s)
- Haiyan Jiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Hongjun Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Zhiyi Yan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Mengbai Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Youming Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Yueyun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Zhe Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Qingyu Ma
- Formula-Pattern Research center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Xiaojuan Li
- Formula-Pattern Research center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Jiaxu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.,Formula-Pattern Research center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, People's Republic of China
| |
Collapse
|
353
|
Genome-Wide Analysis of LysM-Containing Gene Family in Wheat: Structural and Phylogenetic Analysis during Development and Defense. Genes (Basel) 2020; 12:genes12010031. [PMID: 33383636 PMCID: PMC7823900 DOI: 10.3390/genes12010031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 11/17/2022] Open
Abstract
The lysin motif (LysM) family comprise a number of defense proteins that play important roles in plant immunity. The LysM family includes LysM-containing receptor-like proteins (LYP) and LysM-containing receptor-like kinase (LYK). LysM generally recognizes the chitin and peptidoglycan derived from bacteria and fungi. Approximately 4000 proteins with the lysin motif (Pfam PF01476) are found in prokaryotes and eukaryotes. Our study identified 57 LysM genes and 60 LysM proteins in wheat and renamed these genes and proteins based on chromosome distribution. According to the phylogenetic and gene structure of intron-exon distribution analysis, the 60 LysM proteins were classified into seven groups. Gene duplication events had occurred among the LysM family members during the evolution process, resulting in an increase in the LysM gene family. Synteny analysis suggested the characteristics of evolution of the LysM family in wheat and other species. Systematic analysis of these species provided a foundation of LysM genes in crop defense. A comprehensive analysis of the expression and cis-elements of LysM gene family members suggested that they play an essential role in defending against plant pathogens. The present study provides an overview of the LysM family in the wheat genome as well as information on systematic, phylogenetic, gene duplication, and intron-exon distribution analyses that will be helpful for future functional analysis of this important protein family, especially in Gramineae species.
Collapse
|
354
|
Proteomic signatures of 16 major types of human cancer reveal universal and cancer-type-specific proteins for the identification of potential therapeutic targets. J Hematol Oncol 2020; 13:170. [PMID: 33287876 PMCID: PMC7720039 DOI: 10.1186/s13045-020-01013-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/26/2020] [Indexed: 11/10/2022] Open
Abstract
Background Proteomic characterization of cancers is essential for a comprehensive understanding of key molecular aberrations. However, proteomic profiling of a large cohort of cancer tissues is often limited by the conventional approaches. Methods We present a proteomic landscape of 16 major types of human cancer, based on the analysis of 126 treatment-naïve primary tumor tissues, 94 tumor-matched normal adjacent tissues, and 12 normal tissues, using mass spectrometry-based data-independent acquisition approach.
Results In our study, a total of 8527 proteins were mapped to brain, head and neck, breast, lung (both small cell and non-small cell lung cancers), esophagus, stomach, pancreas, liver, colon, kidney, bladder, prostate, uterus and ovary cancers, including 2458 tissue-enriched proteins. Our DIA-based proteomic approach has characterized major human cancers and identified universally expressed proteins as well as tissue-type-specific and cancer-type-specific proteins. In addition, 1139 therapeutic targetable proteins and 21 cancer/testis (CT) antigens were observed. Conclusions Our discoveries not only advance our understanding of human cancers, but also have implications for the design of future large-scale cancer proteomic studies to assist the development of diagnostic and/or therapeutic targets in multiple cancers.
Collapse
|
355
|
Song Y, Yang J, Jing W, Wang Q, Liu Y, Cheng X, Ye F, Tian J, Wei F, Ma S. Systemic elucidation on the potential bioactive compounds and hypoglycemic mechanism of Polygonum multiflorum based on network pharmacology. Chin Med 2020; 15:121. [PMID: 33292335 PMCID: PMC7672844 DOI: 10.1186/s13020-020-00401-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/06/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Diabetes is a complex metabolic disease characterized by hyperglycemia, plaguing the whole world. However, the action mode of multi-component and multi-target for traditional Chinese medicine (TCM) could be a promising treatment of diabetes mellitus. According to the previous research, the TCM of Polygonum multiflorum (PM) showed noteworthy hypoglycemic effect. Up to now, its hypoglycemic active ingredients and mechanism of action are not yet clear. In this study, network pharmacology was employed to elucidate the potential bioactive compounds and hypoglycemic mechanism of PM. METHODS First, the compounds with good pharmacokinetic properties were screened from the self-established library of PM, and the targets of these compounds were predicted and collected through database. Relevant targets of diabetes were summarized by searching database. The intersection targets of compound-targets and disease-targets were obtained soon. Secondly, the interaction net between the compounds and the filtered targets was established. These key targets were enriched and analyzed by protein-protein interactions (PPI) analysis, molecular docking verification. Thirdly, the key genes were used to find the biologic pathway and explain the therapeutic mechanism by genome ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) analysis. Lastly, the part of potential bioactive compounds were under enzyme activity inhibition tests. RESULTS In this study, 29 hypoglycemic components and 63 hypoglycemic targets of PM were filtrated based on online network database. Then the component-target interaction network was constructed and five key components resveratrol, apigenin, kaempferol, quercetin and luteolin were further obtained. Sequential studies turned out, AKT1, EGFR, ESR1, PTGS2, MMP9, MAPK14, and KDR were the common key targets. Docking studies indicated that the bioactive compounds could stably bind the pockets of target proteins. There were 38 metabolic pathways, including regulation of lipolysis in adipocytes, prolactin signaling pathway, TNF signaling pathway, VEGF signaling pathway, FoxO signaling pathway, estrogen signaling pathway, linoleic acid metabolism, Rap1 signaling pathway, arachidonic acid metabolism, and osteoclast differentiation closely connected with the hypoglycemic mechanism of PM. And the enzyme activity inhibition tests showed the bioactive ingredients have great hypoglycemic activity. CONCLUSION In summary, the study used systems pharmacology to elucidate the main hypoglycemic components and mechanism of PM. The work provided a scientific basis for the further hypoglycemic effect research of PM and its monomer components, but also provided a reference for the secondary development of PM.
Collapse
Affiliation(s)
- Yunfei Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Jianbo Yang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Wenguang Jing
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Qi Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Yue Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xianlong Cheng
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Fei Ye
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jinying Tian
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Feng Wei
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China.
| | - Shuangcheng Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China.
| |
Collapse
|
356
|
Verstraete N, Jurman G, Bertagnolli G, Ghavasieh A, Pancaldi V, De Domenico M. CovMulNet19, Integrating Proteins, Diseases, Drugs, and Symptoms: A Network Medicine Approach to COVID-19. NETWORK AND SYSTEMS MEDICINE 2020; 3:130-141. [PMID: 33274348 PMCID: PMC7703682 DOI: 10.1089/nsm.2020.0011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction: We introduce in this study CovMulNet19, a comprehensive COVID-19 network containing all available known interactions involving SARS-CoV-2 proteins, interacting-human proteins, diseases and symptoms that are related to these human proteins, and compounds that can potentially target them. Materials and Methods: Extensive network analysis methods, based on a bootstrap approach, allow us to prioritize a list of diseases that display a high similarity to COVID-19 and a list of drugs that could potentially be beneficial to treat patients. As a key feature of CovMulNet19, the inclusion of symptoms allows a deeper characterization of the disease pathology, representing a useful proxy for COVID-19-related molecular processes. Results: We recapitulate many of the known symptoms of the disease and we find the most similar diseases to COVID-19 reflect conditions that are risk factors in patients. In particular, the comparison between CovMulNet19 and randomized networks recovers many of the known associated comorbidities that are important risk factors for COVID-19 patients, through identified similarities with intestinal, hepatic, and neurological diseases as well as with respiratory conditions, in line with reported comorbidities. Conclusion: CovMulNet19 can be suitably used for network medicine analysis, as a valuable tool for exploring drug repurposing while accounting for the intervening multidimensional factors, from molecular interactions to symptoms.
Collapse
Affiliation(s)
- Nina Verstraete
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 Inserm, ERL5294 CNRS, Toulouse, France
- University Paul Sabatier III, Toulouse, France
| | | | | | | | - Vera Pancaldi
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 Inserm, ERL5294 CNRS, Toulouse, France
- University Paul Sabatier III, Toulouse, France
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
| | | |
Collapse
|
357
|
Liang B, Zhang XX, Gu N. Virtual screening and network pharmacology-based synergistic mechanism identification of multiple components contained in Guanxin V against coronary artery disease. BMC Complement Med Ther 2020; 20:345. [PMID: 33187508 PMCID: PMC7664106 DOI: 10.1186/s12906-020-03133-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Guanxin V (GXV), a traditional Chinese medicine (TCM), has been widely used to treat coronary artery disease (CAD) in clinical practice in China. However, research on the active components and underlying mechanisms of GXV in CAD is still scarce. METHODS A virtual screening and network pharmacological approach was utilized for predicting the pharmacological mechanisms of GXV in CAD. The active compounds of GXV based on various TCM-related databases were selected and then the potential targets of these compounds were identified. Then, after the CAD targets were built through nine databases, a PPI network was constructed based on the matching GXV and CAD potential targets, and the hub targets were screened by MCODE. Moreover, Metascape was applied to GO and KEGG functional enrichment. Finally, HPLC fingerprints of GXV were established. RESULTS A total of 119 active components and 121 potential targets shared between CAD and GXV were obtained. The results of functional enrichment indicated that several GO biological processes and KEGG pathways of GXV mostly participated in the therapeutic mechanisms. Furthermore, 7 hub MCODEs of GXV were collected as potential targets, implying the complex effects of GXV-mediated protection against CAD. Six specific chemicals were identified. CONCLUSION GXV could be employed for CAD through molecular mechanisms, involving complex interactions between multiple compounds and targets, as predicted by virtual screening and network pharmacology. Our study provides a new TCM for the treatment of CAD and deepens the understanding of the molecular mechanisms of GXV against CAD.
Collapse
Affiliation(s)
- Bo Liang
- Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Ning Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
358
|
Sun X, Jiang J, Wang Y, Liu S. Exploring the potential therapeutic effect of traditional Chinese medicine on coronavirus disease 2019 (COVID-19) through a combination of data mining and network pharmacology analysis. Eur J Integr Med 2020; 40:101242. [PMID: 33163124 PMCID: PMC7598573 DOI: 10.1016/j.eujim.2020.101242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 01/18/2023]
Abstract
Introduction Historically traditional Chinese medicine (TCM) has been used as treatment during epidemics. During the recent COVID-19 pandemic patients evidence suggests that the use of TCM has provided health benefits and has been successfully used to control the spread of the disease in China. The aim of this study was to systematically explore the TCM formulae which have been used for the prevention and treatment of pneumonia or ‘pestilence’ to investigate their compatibility with the Chinese materia medica (CMM) and understand their potential mechanisms in the treatment of COVID-19. Methods Frequency analysis was performed to identify high-frequency CMM and CMM groups. Association rules analysis was applied to investigate the compatibility law of CMMs and generate the commonly used CMM groups. Results A total of 173 prescriptions were collected. The frequency analysis showed that seven out of ten high-frequency CMMs overlapped with Lianhua Qingwen Capsules (LHQWC), and five high-frequency pair-CMMs and four triple-CMMs were included in LHQWC, respectively. Then three groups of CMM were generated from association rules analysis, one of which is Ma Xing Shi Gan Decoction (MXSGD). The results of the protein-protein interaction network and enrichment analysis showed that the potential therapeutic mechanisms of the generated prescriptions were involved in the anti-inflammatory, anti-viral, and neuroprotective effects. Conclusion This study showed the importance of systematic research on TCM prescriptions and provided candidate CMM groups that have the potential to treat COVID-19. In vitro and in vivo experiments should be conducted to validate these network pharmacology results, which can provide more information for the development of potential antiviral drugs from TCM prescriptions. The combination of TCM treatment and modern medical approaches will benefit patients with COVID-19 and help to overcome the current epidemic.
Collapse
Affiliation(s)
- Xiuli Sun
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jinhe Jiang
- School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, China
| | - Yang Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China.,State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Shuying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
359
|
Kuenzi BM, Park J, Fong SH, Sanchez KS, Lee J, Kreisberg JF, Ma J, Ideker T. Predicting Drug Response and Synergy Using a Deep Learning Model of Human Cancer Cells. Cancer Cell 2020; 38:672-684.e6. [PMID: 33096023 PMCID: PMC7737474 DOI: 10.1016/j.ccell.2020.09.014] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/07/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022]
Abstract
Most drugs entering clinical trials fail, often related to an incomplete understanding of the mechanisms governing drug response. Machine learning techniques hold immense promise for better drug response predictions, but most have not reached clinical practice due to their lack of interpretability and their focus on monotherapies. We address these challenges by developing DrugCell, an interpretable deep learning model of human cancer cells trained on the responses of 1,235 tumor cell lines to 684 drugs. Tumor genotypes induce states in cellular subsystems that are integrated with drug structure to predict response to therapy and, simultaneously, learn biological mechanisms underlying the drug response. DrugCell predictions are accurate in cell lines and also stratify clinical outcomes. Analysis of DrugCell mechanisms leads directly to the design of synergistic drug combinations, which we validate systematically by combinatorial CRISPR, drug-drug screening in vitro, and patient-derived xenografts. DrugCell provides a blueprint for constructing interpretable models for predictive medicine.
Collapse
Affiliation(s)
- Brent M Kuenzi
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jisoo Park
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Samson H Fong
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kyle S Sanchez
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - John Lee
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jason F Kreisberg
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jianzhu Ma
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
360
|
Matschinske J, Salgado-Albarrán M, Sadegh S, Bongiovanni D, Baumbach J, Blumenthal DB. Individuating Possibly Repurposable Drugs and Drug Targets for COVID-19 Treatment Through Hypothesis-Driven Systems Medicine Using CoVex. Assay Drug Dev Technol 2020; 18:348-355. [PMID: 33164550 PMCID: PMC7703307 DOI: 10.1089/adt.2020.1010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the SARS-CoV-2 virus, has developed into a pandemic causing major disruptions and hundreds of thousands of deaths in wide parts of the world. As of July 3, 2020, neither vaccines nor approved drugs for effective treatment are available. In this article, we showcase how to individuate drug targets and potentially repurposable drugs in silico using CoVex a recently presented systems medicine platform for COVID-19 drug repurposing. Starting from initial hypotheses, CoVex leverages network algorithms to individuate host proteins involved in COVID-19 disease mechanisms, as well as existing drugs targeting these potential drug targets. Our analysis reveals GLA, PLAT, and GGCX as potential drug targets, and urokinase, argatroban, dabigatran etexilate, betrixaban, ximelagatran and anisindione as potentially repurposable drugs.
Collapse
Affiliation(s)
- Julian Matschinske
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| | - Marisol Salgado-Albarrán
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
- Natural Sciences Department, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico
| | - Sepideh Sadegh
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| | - Dario Bongiovanni
- Department of Internal Medicine I, School of Medicine, University Hospital Rechts der Isar, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS, Humanitas University, Rozzano, Milan, Italy
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| | - David B. Blumenthal
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| |
Collapse
|
361
|
Hou R, Wu J, Xu L, Zou Q, Wu YJ. Computational Prediction of Protein Arginine Methylation Based on Composition-Transition-Distribution Features. ACS OMEGA 2020; 5:27470-27479. [PMID: 33134710 PMCID: PMC7594152 DOI: 10.1021/acsomega.0c03972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Arginine methylation is one of the most essential protein post-translational modifications. Identifying the site of arginine methylation is a critical problem in biology research. Unfortunately, biological experiments such as mass spectrometry are expensive and time-consuming. Hence, predicting arginine methylation by machine learning is an alternative fast and efficient way. In this paper, we focus on the systematic characterization of arginine methylation with composition-transition-distribution (CTD) features. The presented framework consists of three stages. In the first stage, we extract CTD features from 1750 samples and exploit decision tree to generate accurate prediction. The accuracy of prediction can reach 96%. In the second stage, the support vector machine can predict the number of arginine methylation sites with 0.36 R-squared. In the third stage, experiments carried out with the updated arginine methylation site data set show that utilizing CTD features and adopting random forest as the classifier outperform previous methods. The accuracy of identification can reach 82.1 and 82.5% in single methylarginine and double methylarginine data sets, respectively. The discovery presented in this paper can be helpful for future research on arginine methylation.
Collapse
Affiliation(s)
- Ruiyan Hou
- Laboratory
of Molecular Toxicology, State Key Laboratory of Integrated Management
of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Science, University of Chinese Academy
of Sciences, Beijing 100049, China
| | - Jin Wu
- School
of Management, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Lei Xu
- School
of Electronic and Engineering, Shenzhen
Polytechnic, Shenzhen 518055, China
| | - Quan Zou
- Institute
of Fundamental and Frontier Sciences, University
of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yi-Jun Wu
- Laboratory
of Molecular Toxicology, State Key Laboratory of Integrated Management
of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
362
|
Feng Z, Chen M, Shen M, Liang T, Chen H, Xie XQ. Pain-CKB, A Pain-Domain-Specific Chemogenomics Knowledgebase for Target Identification and Systems Pharmacology Research. J Chem Inf Model 2020; 60:4429-4435. [PMID: 32786694 DOI: 10.1021/acs.jcim.0c00633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A traditional single-target analgesic, though it may be highly selective and potent, may not be sufficient to mitigate pain. An alternative strategy for alleviation of pain is to seek simultaneous modulation at multiple nodes in the network of pain-signaling pathways through a multitarget analgesic or drug combinations. Here we present a comprehensive pain-domain-specific chemogenomics knowledgebase (Pain-CKB) with integrated computing tools for target identification and systems pharmacology research. Pain-CKB is constructed on the basis of our established chemogenomics technology with new features, including multiple compound support, multicavity protein support, and customizable symbol display. The determination of bioactivity is also revised to avoid the use of complex machine learning models. Our one-stop computing platform describes the chemical molecules, genes, and proteins involved in pain regulation. To date, Pain-CKB has archived 272 analgesics in the market, 84 pain-related targets with 207 available 3D crystal or cryo-EM structures, and 234 662 chemical agents reported for these target proteins. Moreover, Pain-CKB implements user-friendly web-interfaced computing tools and applications for the prediction and analysis of the relevant protein targets and visualization of the outputs, including HTDocking, TargetHunter, BBB permeation predictor, NGL viewer, Spider Plot, etc. The Pain-CKB server is accessible at https://www.cbligand.org/g/pain-ckb.
Collapse
Affiliation(s)
- Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Maozi Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Mingzhe Shen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Tianjian Liang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Hui Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; National Center of Excellence for Computational Drug Abuse Research; Drug Discovery Institute; and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
363
|
Guo Z, Wang P, Liu Z, Zhao Y. Discrimination of Thermophilic Proteins and Non-thermophilic Proteins Using Feature Dimension Reduction. Front Bioeng Biotechnol 2020; 8:584807. [PMID: 33195148 PMCID: PMC7642589 DOI: 10.3389/fbioe.2020.584807] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/11/2020] [Indexed: 01/19/2023] Open
Abstract
Thermophilicity is a very important property of proteins, as it sometimes determines denaturation and cell death. Thus, methods for predicting thermophilic proteins and non-thermophilic proteins are of interest and can contribute to the design and engineering of proteins. In this article, we describe the use of feature dimension reduction technology and LIBSVM to identify thermophilic proteins. The highest accuracy obtained by cross-validation was 96.02% with 119 parameters. When using only 16 features, we obtained an accuracy of 93.33%. We discuss the importance of the different characteristics in identification and report a comparison of the performance of support vector machine to that of other methods.
Collapse
Affiliation(s)
- Zifan Guo
- School of Aeronautics and Astronautic, Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Pingping Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhendong Liu
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Yuming Zhao
- Information and Computer Engineering College, Northeast Forestry University, Harbin, China
| |
Collapse
|
364
|
A Method for Identifying Vesicle Transport Proteins Based on LibSVM and MRMD. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:8926750. [PMID: 33133228 PMCID: PMC7591939 DOI: 10.1155/2020/8926750] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/14/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
With the development of computer technology, many machine learning algorithms have been applied to the field of biology, forming the discipline of bioinformatics. Protein function prediction is a classic research topic in this subject area. Though many scholars have made achievements in identifying protein by different algorithms, they often extract a large number of feature types and use very complex classification methods to obtain little improvement in the classification effect, and this process is very time-consuming. In this research, we attempt to utilize as few features as possible to classify vesicular transportation proteins and to simultaneously obtain a comparative satisfactory classification result. We adopt CTDC which is a submethod of the method of composition, transition, and distribution (CTD) to extract only 39 features from each sequence, and LibSVM is used as the classification method. We use the SMOTE method to deal with the problem of dataset imbalance. There are 11619 protein sequences in our dataset. We selected 4428 sequences to train our classification model and selected other 1832 sequences from our dataset to test the classification effect and finally achieved an accuracy of 71.77%. After dimension reduction by MRMD, the accuracy is 72.16%.
Collapse
|
365
|
Li Q, Xu L, Li Q, Zhang L. Identification and Classification of Enhancers Using Dimension Reduction Technique and Recurrent Neural Network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:8852258. [PMID: 33133227 PMCID: PMC7591959 DOI: 10.1155/2020/8852258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022]
Abstract
Enhancers are noncoding fragments in DNA sequences, which play an important role in gene transcription and translation. However, due to their high free scattering and positional variability, the identification and classification of enhancers have a higher level of complexity than those of coding genes. In order to solve this problem, many computer studies have been carried out in this field, but there are still some deficiencies in these prediction models. In this paper, we use various feature extraction strategies, dimension reduction technology, and a comprehensive application of machine model and recurrent neural network model to achieve an accurate prediction of enhancer identification and classification with the accuracy of was 76.7% and 84.9%, respectively. The model proposed in this paper is superior to the previous methods in performance index or feature dimension, which provides inspiration for the prediction of enhancers by computer technology in the future.
Collapse
Affiliation(s)
- Qingwen Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Qingyuan Li
- Forestry and Fruit Tree Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Lichao Zhang
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen, China
| |
Collapse
|
366
|
Potential Molecular Mechanisms of Chaihu-Shugan-San in Treatment of Breast Cancer Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3670309. [PMID: 33062007 PMCID: PMC7533014 DOI: 10.1155/2020/3670309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Breast cancer is one of the most common cancers endangering women's health all over the world. Traditional Chinese medicine is increasingly recognized as a possible complementary and alternative therapy for breast cancer. Chaihu-Shugan-San is a traditional Chinese medicine prescription, which is extensively used in clinical practice. Its therapeutic effect on breast cancer has attracted extensive attention, but its mechanism of action is still unclear. In this study, we explored the molecular mechanism of Chaihu-Shugan-San in the treatment of breast cancer by network pharmacology. The results showed that 157 active ingredients and 8074 potential drug targets were obtained in the TCMSP database according to the screening conditions. 2384 disease targets were collected in the TTD, OMIM, DrugBank, GeneCards disease database. We applied the Bisogenet plug-in in Cytoscape 3.7.1 to obtain 451 core targets. The biological process of gene ontology (GO) involves the mRNA catabolic process, RNA catabolic process, telomere organization, nucleobase-containing compound catabolic process, heterocycle catabolic process, and so on. In cellular component, cytosolic part, focal adhesion, cell-substrate adherens junction, and cell-substrate junction are highly correlated with breast cancer. In the molecular function category, most proteins were addressed to ubiquitin-like protein ligase binding, protein domain specific binding, and Nop56p-associated pre-rRNA complex. Besides, the results of the KEGG pathway analysis showed that the pathways mainly involved in apoptosis, cell cycle, transcriptional dysregulation, endocrine resistance, and viral infection. In conclusion, the treatment of breast cancer by Chaihu-Shugan-San is the result of multicomponent, multitarget, and multipathway interaction. This study provides a certain theoretical basis for the treatment of breast cancer by Chaihu-Shugan-San and has certain reference value for the development and application of new drugs.
Collapse
|
367
|
Lu Y, Sun J, Hu M, Kong X, Zhong W, Li C. Network Pharmacology Analysis to Uncover the Potential Mechanisms of Lycium barbarum on Colorectal Cancer. Interdiscip Sci 2020; 12:515-525. [PMID: 33048277 DOI: 10.1007/s12539-020-00397-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/01/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Studies have shown that extracts from Lycium barbarum exerted protective effects against colorectal cancer (CRC) cells. We used the network pharmacology method to determine the effects of L. barbarum on CRC and to predict core targets, biological functions, pathways, and mechanisms of action. METHOD We obtained the active compounds and their targets in L. barbarum via use of the Traditional Chinese Medicine System Pharmacology Database (TCMSP), gathered the CRC targets from Malacards, TTD, GeneCards, and DisGeNET, and chosen the overlapped targets as the candidate targets. After protein-protein interaction (PPI) network analysis, 20 with the highest node degree were selected as the core targets, and their enrichment and pathways were analyzed. Furthermore, we employed iGEMDOCK to validate the compound-target relation. RESULT Eventually, 103 overlapped targets were chosen as the candidate targets. Targets with the top 20 highest node degree were selected as the core targets. Gene Ontology (GO) enrichment analysis indicated that the core targets were enriched in cell proliferation regulation, extracellular space, cytokine receptor binding, and so on. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis proved that the core targets were significantly enriched in bladder cancer, pathways in cancer. The docking results demonstrated that beta-sitosterol, glycitein, and quercetin had good binding activity to CRC putative targets. CONCLUSION Our work successfully predicted the functioning ingredients and potential targets of L. barbarum in CRC and illustrated the potential pathways and mechanisms comprehensively. Nevertheless, these results still call for in vitro and in vivo experiments to validate.
Collapse
Affiliation(s)
- Yi Lu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiachen Sun
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Minhui Hu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xianhe Kong
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weijie Zhong
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chujun Li
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China. .,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
368
|
Computer-Aided Estimation of Biological Activity Profiles of Drug-Like Compounds Taking into Account Their Metabolism in Human Body. Int J Mol Sci 2020; 21:ijms21207492. [PMID: 33050610 PMCID: PMC7593915 DOI: 10.3390/ijms21207492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/25/2022] Open
Abstract
Most pharmaceutical substances interact with several or even many molecular targets in the organism, determining the complex profiles of their biological activity. Moreover, due to biotransformation in the human body, they form one or several metabolites with different biological activity profiles. Therefore, the development and rational use of novel drugs requires the analysis of their biological activity profiles, taking into account metabolism in the human body. In silico methods are currently widely used for estimating new drug-like compounds' interactions with pharmacological targets and predicting their metabolic transformations. In this study, we consider the estimation of the biological activity profiles of organic compounds, taking into account the action of both the parent molecule and its metabolites in the human body. We used an external dataset that consists of 864 parent compounds with known metabolites. It is shown that the complex assessment of active pharmaceutical ingredients' interactions with the human organism increases the quality of computer-aided estimates. The toxic and adverse effects showed the most significant difference: reaching 0.16 for recall and 0.14 for precision.
Collapse
|
369
|
Zhang Y, Zhang X, Zhang X, Cai Y, Cheng M, Yan C, Han Y. Molecular Targets and Pathways Contributing to the Effects of Wenxin Keli on Atrial Fibrillation Based on a Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:8396484. [PMID: 33123211 PMCID: PMC7586041 DOI: 10.1155/2020/8396484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common sustained arrhythmia and is associated with high rates of mortality and morbidity. The traditional Chinese medicine Wenxin Keli (WXKL) can effectively improve clinical symptoms and is safe for the treatment of AF. However, the active substances in WXKL and the molecular mechanisms underlying its effects on AF remain unclear. In this study, the bioactive compounds in WXKL, as well as their molecular targets and associated pathways, were evaluated by systems pharmacology. MATERIALS AND METHODS Chemical constituents and potential targets of WXKL were obtained via the Traditional Chinese Medicine Systems Pharmacology (TCMSP). The TTD, DrugBank, DisGeNET, and GeneCards databases were used to collect AF-related target genes. Based on common targets related to both AF and WXKL, a protein interaction network was generated using the STRING database. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGGs) pathway enrichment analyses were performed. Network diagrams of the active component-target and protein-protein interactions (PPIs) were constructed using Cytoscape. RESULTS A total of 30 active ingredients in WXKL and 219 putative target genes were screened, including 83 genes identified as therapeutic targets in AF; these overlapping genes were considered candidate targets for subsequent analyses. The effect of treating AF was mainly correlated with the regulation of target proteins, such as IL-6, TNF, AKT1, VEGFA, CXCL8, TP53, CCL2, MMP9, CASP3, and NOS3. GO and KEGG analyses revealed that these targets are associated with the inflammatory response, oxidative stress reaction, immune regulation, cardiac energy metabolism, serotonergic synapse, and other pathways. CONCLUSIONS This study demonstrated the multicomponent, multitarget, and multichannel characteristics of WXKL, providing a basis for further studies of the mechanism underlying the beneficial effects of WXKL in AF.
Collapse
Affiliation(s)
- Yujie Zhang
- Liaoning University of Traditional Chinese Medicine, Liaoning, Shenyang 110847, China
| | - Xiaolin Zhang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theatre Command, Liaoning, Shenyang 110840, China
| | - Xi Zhang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theatre Command, Liaoning, Shenyang 110840, China
| | - Yi Cai
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theatre Command, Liaoning, Shenyang 110840, China
| | - Minghui Cheng
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theatre Command, Liaoning, Shenyang 110840, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theatre Command, Liaoning, Shenyang 110840, China
| | - Yaling Han
- Liaoning University of Traditional Chinese Medicine, Liaoning, Shenyang 110847, China
| |
Collapse
|
370
|
Uncovering the protective mechanism of Taohong Siwu decoction against diabetic retinopathy via HIF-1 signaling pathway based on network analysis and experimental validation. BMC Complement Med Ther 2020; 20:298. [PMID: 33023593 PMCID: PMC7542117 DOI: 10.1186/s12906-020-03086-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background Diabetic retinopathy (DR) is a common and serious microvascular complication of diabetes. Taohong Siwu decoction (THSWD), a famous traditional Chinese medicine (TCM) prescription, has been proved to have a good clinical effect on DR, whereas its molecular mechanism remains unclear. Our study aimed to uncover the core targets and signaling pathways of THSWD against DR. Methods First, the active ingredients of THSWD were searched from Traditional Chinese Medicine Systems Pharmacology (TCMSP) Database. Second, the targets of active ingredients were identified from ChemMapper and PharmMapper databases. Third, DR associated targets were searched from DisGeNET, DrugBank and Therapeutic Target Database (TTD). Subsequently, the common targets of active ingredients and DR were found and analyzed in STRING database. DAVID database and ClueGo plug-in software were used to carry out the gene ontology (GO) and KEGG enrichment analysis. The core signaling pathway network of “herb-ingredient-target” was constructed by the Cytoscape software. Finally, the key genes of THSWD against DR were validated by quantitative real-time PCR (qRT-PCR). Results A total of 2340 targets of 61 active ingredients in THSWD were obtained. Simultaneously, a total of 263 DR-associated targets were also obtained. Then, 67 common targets were found by overlapping them, and 23 core targets were identified from protein-protein interaction (PPI) network. Response to hypoxia was found as the top GO term of biological process, and HIF-1 signaling pathway was found as the top KEGG pathway. Among the key genes in HIF-1 pathway, the mRNA expression levels of VEGFA, SERPINE1 and NOS2 were significantly down-regulated by THSWD (P < 0.05), and NOS3 and HMOX1 were significantly up-regulated (P < 0.05). Conclusion THSWD had a protective effect on DR via regulating HIF-1 signaling pathway and other important pathways. This study might provide a theoretical basis for the application of THSWD and the development of new drugs for the treatment of DR.
Collapse
|
371
|
A Network Pharmacology Technique to Investigate the Synergistic Mechanisms of Salvia miltiorrhiza and Radix puerariae in Treatment of Cardio-Cerebral Vascular Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6937186. [PMID: 33082828 PMCID: PMC7566220 DOI: 10.1155/2020/6937186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022]
Abstract
Objective This study is aimed to analyze the active ingredients, drug targets, and related pathways in the combination of Salvia miltiorrhiza (SM) and Radix puerariae (RP) in the treatment of cardio-cerebral vascular diseases (CCVDs). Method The ingredients and targets of SM and RP were obtained from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), and the disease targets were obtained from Therapeutic Target Database (TTD), National Center for Biotechnology Information (NCBI), and Online Mendelian Inheritance in Man (OMIM) Database. The synergistic mechanisms of the SM and RP were evaluated by gene ontology (GO) enrichment analyses and Kyoto encyclopedia of genes and genomes (KEGG) path enrichment analyses. Result A total of 61 active ingredients and 58 common targets were identified in this study. KEGG pathway enrichment analysis results showed that SM- and RP-regulated pathways were mainly inflammatory processes, immunosuppression, and cardiovascular systems. The component-target-pathway network indicated that SM and RP exert a synergistic mechanism for CCVDs through PTGS2 target in PI3k-Akt, TNF, and Jak-STAT signaling pathways. Conclusion In summary, this study clarified the synergistic mechanisms of SM and RP, which can provide a better understanding of effect in the treatment of CCVDs.
Collapse
|
372
|
Mi B, Li Q, Li T, Marshall J, Sai J. A network pharmacology study on analgesic mechanism of Yuanhu-Baizhi herb pair. BMC Complement Med Ther 2020; 20:284. [PMID: 32948176 PMCID: PMC7501664 DOI: 10.1186/s12906-020-03078-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/13/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Millions of people are suffering from chronic pain conditions, such as headache, arthritis, cancer. Apart from western medicines, traditional Chinese medicines are also well accepted for pain management, especially in Asian countries. Yuanhu-Baizhi herb pair (YB) is a typical herb pair applied to the treatment of stomach pain, hypochondriac pain, headache, and dysmenorrhea, due to its effects on analgesia and sedation. This study is to identify potentially active compounds and the underlying mechanisms of YB in the treatment of pain. METHODS Compounds in YB were collected from 3 online databases and then screened by bioavailability and drug likeness parameters. Swiss target prediction was applied to obtain targets information of the active compounds. Pain-related genes were conducted for Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Protein-protein interaction (PPI) networks of the genes were constructed using Cytoscape software. In addition, the hub genes were screened using maximal clique centrality (MCC) algorithm. RESULTS In total, 31 compounds from Yuanhu were screened out with 35 putative target genes, while 26 compounds in Baizhi with 43 target genes were discovered. Hence, 78 potential target genes of YB were selected for further study. After overlap analysis of the 78 genes of YB and 2408 pain-associated genes, we finally achieved 34 YB-pain target genes, as well as 10 hub genes and 23 core compounds. Go enrichment and KEGG pathway analysis indicated that YB had a strong integration with neuro system, which might significantly contribute to antinociceptive effect. CONCLUSION Our data provide deep understanding of the pharmacological mechanisms of YB in attenuating pain. The discovery shed new light on the development of active compounds of YB for the treatment of pain.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiushi Li
- Department of Cardiology, Beijing Chaoyang Integrative Medicine Emergency Medical Center, Beijing, 100029, China
| | - Tong Li
- Department of oncology, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jessica Marshall
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, Boston, MA, 02115, USA
| | - Jiayang Sai
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Department of oncology, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
373
|
Essentiality and Transcriptome-Enriched Pathway Scores Predict Drug-Combination Synergy. BIOLOGY 2020; 9:biology9090278. [PMID: 32906805 PMCID: PMC7565142 DOI: 10.3390/biology9090278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022]
Abstract
In the prediction of the synergy of drug combinations, systems pharmacology models expand the scope of experiment screening and overcome the limitations of current computational models posed by their lack of mechanical interpretation and integration of gene essentiality. We therefore investigated the synergy of drug combinations for cancer therapies utilizing records in NCI ALMANAC, and we employed logistic regression to test the statistical significance of gene and pathway features in that interaction. We trained our predictive models using 43 NCI-60 cell lines, 165 KEGG pathways, and 114 drug pairs. Scores of drug-combination synergies showed a stronger correlation with pathway than gene features in overall trend analysis and a significant association with both genes and pathways in genome-wide association analyses. However, we observed little overlap of significant gene expressions and essentialities and no significant evidence that associated target and non-target genes and their pathways. We were able to validate four drug-combination pathways between two drug combinations, Nelarabine-Exemestane and Docetaxel-Vermurafenib, and two signaling pathways, PI3K-AKT and AMPK, in 16 cell lines. In conclusion, pathways significantly outperformed genes in predicting drug-combination synergy, and because they have very different mechanisms, gene expression and essentiality should be considered in combination rather than individually to improve this prediction.
Collapse
|
374
|
Chaudhari R, Fong LW, Tan Z, Huang B, Zhang S. An up-to-date overview of computational polypharmacology in modern drug discovery. Expert Opin Drug Discov 2020; 15:1025-1044. [PMID: 32452701 PMCID: PMC7415563 DOI: 10.1080/17460441.2020.1767063] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/06/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION In recent years, computational polypharmacology has gained significant attention to study the promiscuous nature of drugs. Despite tremendous challenges, community-wide efforts have led to a variety of novel approaches for predicting drug polypharmacology. In particular, some rapid advances using machine learning and artificial intelligence have been reported with great success. AREAS COVERED In this article, the authors provide a comprehensive update on the current state-of-the-art polypharmacology approaches and their applications, focusing on those reports published after our 2017 review article. The authors particularly discuss some novel, groundbreaking concepts, and methods that have been developed recently and applied to drug polypharmacology studies. EXPERT OPINION Polypharmacology is evolving and novel concepts are being introduced to counter the current challenges in the field. However, major hurdles remain including incompleteness of high-quality experimental data, lack of in vitro and in vivo assays to characterize multi-targeting agents, shortage of robust computational methods, and challenges to identify the best target combinations and design effective multi-targeting agents. Fortunately, numerous national/international efforts including multi-omics and artificial intelligence initiatives as well as most recent collaborations on addressing the COVID-19 pandemic have shown significant promise to propel the field of polypharmacology forward.
Collapse
Affiliation(s)
- Rajan Chaudhari
- Intelligent Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Long Wolf Fong
- Intelligent Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
- MD Anderson UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Avenue, Houston, Texas 77030, United States
| | - Zhi Tan
- Intelligent Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Beibei Huang
- Intelligent Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Shuxing Zhang
- Intelligent Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
- MD Anderson UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Avenue, Houston, Texas 77030, United States
| |
Collapse
|
375
|
Study on Intervention Mechanism of Yiqi Huayu Jiedu Decoction on ARDS Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4782470. [PMID: 32849901 PMCID: PMC7439204 DOI: 10.1155/2020/4782470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/17/2020] [Accepted: 07/11/2020] [Indexed: 02/05/2023]
Abstract
Background Yiqi Huayu Jiedu (YQHYJD) is a traditional Chinese medicine decoction made up of eight traditional Chinese medicines. Although YQHYJD is effectively used to prevent and treat ARDS/acute lung injury (ALI) in rats, the molecular mechanisms supporting its clinical application remain elusive. The purpose of the current study was to understand its lung protective effects at the molecular level using network pharmacology approach. Methods In an ARDS animal model, the beneficial pharmacological activities of YQHYJD were confirmed by reduced lung tissue damage levels observed on drug treated rats versus control group. We then proposed a network analysis to discover the key nodes based on drugs and disease network. Subsequently, we analyzed interaction networks and screened key targets. Using Western blot to detect the expression level of key targets, the intervention effect of changes in expression level of key targets on ARDS was evaluated. Results Pathway enrichment analysis of highly ranked genes showed that ErbB pathways were highly related to ARDS. Finally, western blot results showed decreased level of the AKT1 and KRAS/NRAS/HRAS protein in the lung after treatment which confirmed the hypothesis. Conclusion In conclusion, our results suggest that YQHYJD can exert lung tissue protective effect against the severe injury through multiple pathways, including the endothelial cells permeability improvement, inflammatory reaction inhibition, edema, and lung tissue hemorrhage reduction.
Collapse
|
376
|
Hu Q, Wei S, Wen J, Zhang W, Jiang Y, Qu C, Xiang J, Zhao Y, Peng X, Ma X. Network pharmacology reveals the multiple mechanisms of Xiaochaihu decoction in the treatment of non-alcoholic fatty liver disease. BioData Min 2020; 13:11. [PMID: 32863886 PMCID: PMC7450930 DOI: 10.1186/s13040-020-00224-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver (NAFLD) is a chronic disease worldwide, which poses a huge threat to human health. Xiaochaihu decoction is a well-known traditional Chinese medicine prescription. It has been proven effective in treating NAFLD but its mechanism is still unclear. OBJECTIVE Multiple mechanisms of Xiaochaihu decoction are explored by identifying and connecting potential targets and active ingredients in the treatment of NAFLD. METHODS Active ingredients and related targets of seven herbs were collected from TCMSP database. The related targets of NAFLD were obtained from Genes cards database, TDD and OMIM database. The intersected targets of disease targets and drug targets were input into STRING database to construct protein-protein interaction network. DAVID database was used for GO enrichment analysis and KEGG enrichment analysis. RESULTS After screening and removal of duplicates, a total of 145 active ingredients and 105 potential targets were obtained. PPI network manifested that AKT1, IL6, JUN MAPK8 and STAT3 were the key target proteins. The results of GO enrichment analysis mainly involved cytokine receptor binding, cytokine activity, and heme binding. The results of KEGG analysis suggested that the mechanism mainly involved in AGE-RAGE signaling pathway in diabetic complications, Hepatitis C, fluid shear stress and atherosclerosis. The signaling pathways were further integrated as network manner, including AGE-RAGE signaling pathway in diabetic complications, Fluid shear stress and atherosclerosis, Insulin resistance, HIF-1 signaling pathway, Th17 cell differentiation and IL-17 signaling pathway. The network contained immunity regulation, metabolism regulation and oxidative stress regulation. CONCLUSION Xiaochaihu decoction plays a key role in the treatment of NAFLD with multiple targets and pathways. Immunity regulation, metabolism regulation and oxidative stress regulation consist of the crucial regulation cores in mechanism. GRAPHICAL ABSTRACT Design and workflow of this study.
Collapse
Affiliation(s)
- Qichao Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Shizhang Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
- Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039 China
| | - Jianxia Wen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
- Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039 China
| | - Wenwen Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Yinxiao Jiang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Caiyan Qu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Junbao Xiang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Yanling Zhao
- Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039 China
| | - Xi Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Xiao Ma
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| |
Collapse
|
377
|
Wang L, Lang GT, Xue MZ, Yang L, Chen L, Yao L, Li XG, Wang P, Hu X, Shao ZM. Dissecting the heterogeneity of the alternative polyadenylation profiles in triple-negative breast cancers. Am J Cancer Res 2020; 10:10531-10547. [PMID: 32929364 PMCID: PMC7482814 DOI: 10.7150/thno.40944] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 08/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive malignancy with high heterogeneity. However, the alternative polyadenylation (APA) profiles of TNBC remain unknown. Here, we aimed to define the characteristics of the APA events at post-transcription level among TNBCs. Methods: Using transcriptome microarray data, we analyzed APA profiles of 165 TNBC samples and 33 paired normal tissues. A pooled short hairpin RNA screen targeting 23 core cleavage and polyadenylation (C/P) genes was used to identify key C/P factors. Results: We established an unconventional APA subtyping system composed of four stable subtypes: 1) luminal androgen receptor (LAR), 2) mesenchymal-like immune-activated (MLIA), 3) basal-like (BL), 4) suppressed (S) subtypes. Patients in the S subtype had the worst disease-free survival comparing to other patients (log-rank p = 0.021). Enriched clinically actionable pathways and putative therapeutic APA events were analyzed among each APA subtype. Furthermore, CPSF1 and PABPN1 were identified as the master C/P factors in regulating APA events and TNBC proliferation. The depletion of CPSF1 or PABPN1 weakened cell proliferation, enhanced apoptosis, resulted in cell cycle redistribution and a reversion of APA events of genes associated with tumorigenesis, proliferation, metastasis and chemosensitivity in breast cancer. Conclusions: Our findings advance the understanding of tumor heterogeneity regulation in APA and yield new insights into therapeutic target identification in TNBC.
Collapse
|
378
|
Cai S, Chen Y, Lin S, Ye C, Zheng F, Dong L. Multiple Processes May Involve in the IgG4-RD Pathogenesis: An Integrative Study via Proteomic and Transcriptomic Analysis. Front Immunol 2020; 11:1795. [PMID: 32973752 PMCID: PMC7468437 DOI: 10.3389/fimmu.2020.01795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/06/2020] [Indexed: 01/13/2023] Open
Abstract
Immunoglobulin G4-related disease (IgG4-RD) is a newly defined disease entity, while the exact pathogenesis is still not clear. Identifying the characters of IgG4-RD in proteomic and transcriptomic aspects will be critical to investigate the potential pathogenic mechanisms of IgG4-RD. We performed proteomic analysis realized with iTRAQ technique for serum samples from eight treatment-naive IgG4-RD patients and eight healthy volunteers, and tissue samples from two IgG4-RD patients and two non-IgG4-RD patients. Transcriptomic data (GSE40568 and GSE66465) was obtained from the GEO Dataset for validation. The weighted correlation network analysis (WGCNA) was applied to detect the gene modules correlated with IgG4-RD. KEGG pathway analysis was used to investigate pathways enriched in IgG4-RD samples. As a result, a total of 980 differentially expressed proteins (DEPs) in tissue and 94 DEPs in serum were identified between IgG4-RD and control groups. Three hundred fifty-four and two hundred forty-seven genes that most correlated with IgG4-RD were detected by WGCNA analysis in tissue and PBMC, respectively. We also found that DEPs in IgG4-RD samples were enriched in several immune-related activities including bacterial/viral infections and platelet activation as well as some immune related signaling pathways. In conclusion, we identified multiple processes/factors and several signaling pathways that may involve in the IgG4-RD pathogenesis, and found out some potential therapeutic targets for IgG4-RD.
Collapse
Affiliation(s)
- Shaozhe Cai
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Chen
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - ShengYan Lin
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Ye
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
379
|
Failli M, Paananen J, Fortino V. ThETA: transcriptome-driven efficacy estimates for gene-based TArget discovery. Bioinformatics 2020; 36:4214-4216. [PMID: 32437556 PMCID: PMC7390989 DOI: 10.1093/bioinformatics/btaa518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/23/2020] [Accepted: 05/16/2020] [Indexed: 01/17/2023] Open
Abstract
Summary Estimating efficacy of gene–target-disease associations is a fundamental step in drug discovery. An important data source for this laborious task is RNA expression, which can provide gene–disease associations on the basis of expression fold change and statistical significance. However, the simply use of the log-fold change can lead to numerous false-positive associations. On the other hand, more sophisticated methods that utilize gene co-expression networks do not consider tissue specificity. Here, we introduce Transcriptome-driven Efficacy estimates for gene-based TArget discovery (ThETA), an R package that enables non-expert users to use novel efficacy scoring methods for drug–target discovery. In particular, ThETA allows users to search for gene perturbation (therapeutics) that reverse disease-gene expression and genes that are closely related to disease-genes in tissue-specific networks. ThETA also provides functions to integrate efficacy evaluations obtained with different approaches and to build an overall efficacy score, which can be used to identify and prioritize gene(target)–disease associations. Finally, ThETA implements visualizations to show tissue-specific interconnections between target and disease-genes, and to indicate biological annotations associated with the top selected genes. Availability and implementation ThETA is freely available for academic use at https://github.com/vittoriofortino84/ThETA. Contact vittorio.fortino@uef.fi Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mario Failli
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland.,Department of Chemical, Materials and Industrial Engineering, University of Naples 'Federico II', Naples 80125, Italy
| | - Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland.,Blueprint Genetics Ltd, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland.,Blueprint Genetics Ltd, Finland
| |
Collapse
|
380
|
Li Q, Zhou W, Wang D, Wang S, Li Q. Prediction of Anticancer Peptides Using a Low-Dimensional Feature Model. Front Bioeng Biotechnol 2020; 8:892. [PMID: 32903381 PMCID: PMC7434836 DOI: 10.3389/fbioe.2020.00892] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/10/2020] [Indexed: 01/09/2023] Open
Abstract
Cancer is still a severe health problem globally. The therapy of cancer traditionally involves the use of radiotherapy or anticancer drugs to kill cancer cells, but these methods are quite expensive and have side effects, which will cause great harm to patients. With the find of anticancer peptides (ACPs), significant progress has been achieved in the therapy of tumors. Therefore, it is invaluable to accurately identify anticancer peptides. Although biochemical experiments can solve this work, this method is expensive and time-consuming. To promote the application of anticancer peptides in cancer therapy, machine learning can be used to recognize anticancer peptides by extracting the feature vectors of anticancer peptides. Nevertheless, poor performance usually be found in training the machine learning model to utilizing high-dimensional features in practice. In order to solve the above job, this paper put forward a 19-dimensional feature model based on anticancer peptide sequences, which has lower dimensionality and better performance than some existing methods. In addition, this paper also separated a model with a low number of dimensions and acceptable performance. The few features identified in this study may represent the important features of anticancer peptides.
Collapse
Affiliation(s)
- Qingwen Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Wenyang Zhou
- Center for Bioinformatics, School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, China
| | - Donghua Wang
- Department of General Surgery, Heilongjiang Province Land Reclamation Headquarters General Hospital, Harbin, China
| | - Sui Wang
- Key Laboratory of Soybean Biology in Chinese Ministry of Education, Northeast Agricultural University, Harbin, China
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
| | - Qingyuan Li
- Forestry and Fruit Tree Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
381
|
Singh N, Chaput L, Villoutreix BO. Fast Rescoring Protocols to Improve the Performance of Structure-Based Virtual Screening Performed on Protein-Protein Interfaces. J Chem Inf Model 2020; 60:3910-3934. [PMID: 32786511 DOI: 10.1021/acs.jcim.0c00545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Protein-protein interactions (PPIs) are attractive targets for drug design because of their essential role in numerous cellular processes and disease pathways. However, in general, PPIs display exposed binding pockets at the interface, and as such, have been largely unexploited for therapeutic interventions with low-molecular weight compounds. Here, we used docking and various rescoring strategies in an attempt to recover PPI inhibitors from a set of active and inactive molecules for 11 targets collected in ChEMBL and PubChem. Our focus is on the screening power of the various developed protocols and on using fast approaches so as to be able to apply such a strategy to the screening of ultralarge libraries in the future. First, we docked compounds into each target using the fast "pscreen" mode of the structure-based virtual screening (VS) package Surflex. Subsequently, the docking poses were postprocessed to derive a set of 3D topological descriptors: (i) shape similarity and (ii) interaction fingerprint similarity with a co-crystallized inhibitor, (iii) solvent-accessible surface area, and (iv) extent of deviation from the geometric center of a reference inhibitor. The derivatized descriptors, together with descriptor-scaled scoring functions, were utilized to investigate possible impacts on VS performance metrics. Moreover, four standalone scoring functions, RF-Score-VS (machine-learning), DLIGAND2 (knowledge-based), Vinardo (empirical), and X-SCORE (empirical), were employed to rescore the PPI compounds. Collectively, the results indicate that the topological scoring algorithms could be valuable both at a global level, with up to 79% increase in areas under the receiver operating characteristic curve for some targets, and in early stages, with up to a 4-fold increase in enrichment factors at 1% of the screened collections. Outstandingly, DLIGAND2 emerged as the best scoring function on this data set, outperforming all rescoring techniques in terms of VS metrics. The described methodology could help in the rational design of small-molecule PPI inhibitors and has direct applications in many therapeutic areas, including cancer, CNS, and infectious diseases such as COVID-19.
Collapse
Affiliation(s)
- Natesh Singh
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Ludovic Chaput
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Bruno O Villoutreix
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| |
Collapse
|
382
|
Jarada TN, Rokne JG, Alhajj R. A review of computational drug repositioning: strategies, approaches, opportunities, challenges, and directions. J Cheminform 2020; 12:46. [PMID: 33431024 PMCID: PMC7374666 DOI: 10.1186/s13321-020-00450-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/13/2020] [Indexed: 01/13/2023] Open
Abstract
Drug repositioning is the process of identifying novel therapeutic potentials for existing drugs and discovering therapies for untreated diseases. Drug repositioning, therefore, plays an important role in optimizing the pre-clinical process of developing novel drugs by saving time and cost compared to the traditional de novo drug discovery processes. Since drug repositioning relies on data for existing drugs and diseases the enormous growth of publicly available large-scale biological, biomedical, and electronic health-related data along with the high-performance computing capabilities have accelerated the development of computational drug repositioning approaches. Multidisciplinary researchers and scientists have carried out numerous attempts, with different degrees of efficiency and success, to computationally study the potential of repositioning drugs to identify alternative drug indications. This study reviews recent advancements in the field of computational drug repositioning. First, we highlight different drug repositioning strategies and provide an overview of frequently used resources. Second, we summarize computational approaches that are extensively used in drug repositioning studies. Third, we present different computing and experimental models to validate computational methods. Fourth, we address prospective opportunities, including a few target areas. Finally, we discuss challenges and limitations encountered in computational drug repositioning and conclude with an outline of further research directions.
Collapse
Affiliation(s)
- Tamer N Jarada
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada
| | - Jon G Rokne
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada
| | - Reda Alhajj
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada.
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
383
|
Tang J, Wang Y, Luo Y, Fu J, Zhang Y, Li Y, Xiao Z, Lou Y, Qiu Y, Zhu F. Computational advances of tumor marker selection and sample classification in cancer proteomics. Comput Struct Biotechnol J 2020; 18:2012-2025. [PMID: 32802273 PMCID: PMC7403885 DOI: 10.1016/j.csbj.2020.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer proteomics has become a powerful technique for characterizing the protein markers driving transformation of malignancy, tracing proteome variation triggered by therapeutics, and discovering the novel targets and drugs for the treatment of oncologic diseases. To facilitate cancer diagnosis/prognosis and accelerate drug target discovery, a variety of methods for tumor marker identification and sample classification have been developed and successfully applied to cancer proteomic studies. This review article describes the most recent advances in those various approaches together with their current applications in cancer-related studies. Firstly, a number of popular feature selection methods are overviewed with objective evaluation on their advantages and disadvantages. Secondly, these methods are grouped into three major classes based on their underlying algorithms. Finally, a variety of sample separation algorithms are discussed. This review provides a comprehensive overview of the advances on tumor maker identification and patients/samples/tissues separations, which could be guidance to the researches in cancer proteomics.
Collapse
Key Words
- ANN, Artificial Neural Network
- ANOVA, Analysis of Variance
- CFS, Correlation-based Feature Selection
- Cancer proteomics
- Computational methods
- DAPC, Discriminant Analysis of Principal Component
- DT, Decision Trees
- EDA, Estimation of Distribution Algorithm
- FC, Fold Change
- GA, Genetic Algorithms
- GR, Gain Ratio
- HC, Hill Climbing
- HCA, Hierarchical Cluster Analysis
- IG, Information Gain
- LDA, Linear Discriminant Analysis
- LIMMA, Linear Models for Microarray Data
- MBF, Markov Blanket Filter
- MWW, Mann–Whitney–Wilcoxon test
- OPLS-DA, Orthogonal Partial Least Squares Discriminant Analysis
- PCA, Principal Component Analysis
- PLS-DA, Partial Least Square Discriminant Analysis
- RF, Random Forest
- RF-RFE, Random Forest with Recursive Feature Elimination
- SA, Simulated Annealing
- SAM, Significance Analysis of Microarrays
- SBE, Sequential Backward Elimination
- SFS, and Sequential Forward Selection
- SOM, Self-organizing Map
- SU, Symmetrical Uncertainty
- SVM, Support Vector Machine
- SVM-RFE, Support Vector Machine with Recursive Feature Elimination
- Sample classification
- Tumor marker selection
- sPLSDA, Sparse Partial Least Squares Discriminant Analysis
- t-SNE, Student t Distribution
- χ2, Chi-square
Collapse
Affiliation(s)
- Jing Tang
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Yi Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziyu Xiao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Yunqing Qiu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Feng Zhu
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
384
|
Ratnakumar A, Weinhold N, Mar JC, Riaz N. Protein-Protein interactions uncover candidate 'core genes' within omnigenic disease networks. PLoS Genet 2020; 16:e1008903. [PMID: 32678846 PMCID: PMC7390454 DOI: 10.1371/journal.pgen.1008903] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 07/29/2020] [Accepted: 06/01/2020] [Indexed: 01/09/2023] Open
Abstract
Genome wide association studies (GWAS) of human diseases have generally identified many loci associated with risk with relatively small effect sizes. The omnigenic model attempts to explain this observation by suggesting that diseases can be thought of as networks, where genes with direct involvement in disease-relevant biological pathways are named ‘core genes’, while peripheral genes influence disease risk via their interactions or regulatory effects on core genes. Here, we demonstrate a method for identifying candidate core genes solely from genes in or near disease-associated SNPs (GWAS hits) in conjunction with protein-protein interaction network data. Applied to 1,381 GWAS studies from 5 ancestries, we identify a total of 1,865 candidate core genes in 343 GWAS studies. Our analysis identifies several well-known disease-related genes that are not identified by GWAS, including BRCA1 in Breast Cancer, Amyloid Precursor Protein (APP) in Alzheimer’s Disease, INS in A1C measurement and Type 2 Diabetes, and PCSK9 in LDL cholesterol, amongst others. Notably candidate core genes are preferentially enriched for disease relevance over GWAS hits and are enriched for both Clinvar pathogenic variants and known drug targets—consistent with the predictions of the omnigenic model. We subsequently use parent term annotations provided by the GWAS catalog, to merge related GWAS studies and identify candidate core genes in over-arching disease processes such as cancer–where we identify 109 candidate core genes. A recent theory suggests that only a small number of genes underpin the biology of a disease, these genes are called ‘core genes’, and for most diseases, these core genes remain unknown. The suggested methods for finding them requires complex and expensive experiments. We reasoned that if we merge currently available datasets in smart ways, we may be able to uncover these ‘core genes’. Our method finds “hub” proteins by merging lists of genes previously linked with disease to information on how proteins interact with each other. We found that many of these hub proteins have central roles in disease, such as insulin for both A1C measurement and Type 2 Diabetes, BRCA1 in Breast cancer, and Amyloid Precursor Protein in Alzheimer’s Disease. We think these ‘hub’ proteins are candidate ‘core genes’, and offer our method as a way to find ‘core genes’ by utilizing publicly available reference datasets.
Collapse
Affiliation(s)
- Abhirami Ratnakumar
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| | - Nils Weinhold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Jessica C. Mar
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| |
Collapse
|
385
|
Sadegh S, Matschinske J, Blumenthal DB, Galindez G, Kacprowski T, List M, Nasirigerdeh R, Oubounyt M, Pichlmair A, Rose TD, Salgado-Albarrán M, Späth J, Stukalov A, Wenke NK, Yuan K, Pauling JK, Baumbach J. Exploring the SARS-CoV-2 virus-host-drug interactome for drug repurposing. Nat Commun 2020; 11:3518. [PMID: 32665542 PMCID: PMC7360763 DOI: 10.1038/s41467-020-17189-2] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/12/2020] [Indexed: 11/09/2022] Open
Abstract
Coronavirus Disease-2019 (COVID-19) is an infectious disease caused by the SARS-CoV-2 virus. Various studies exist about the molecular mechanisms of viral infection. However, such information is spread across many publications and it is very time-consuming to integrate, and exploit. We develop CoVex, an interactive online platform for SARS-CoV-2 host interactome exploration and drug (target) identification. CoVex integrates virus-human protein interactions, human protein-protein interactions, and drug-target interactions. It allows visual exploration of the virus-host interactome and implements systems medicine algorithms for network-based prediction of drug candidates. Thus, CoVex is a resource to understand molecular mechanisms of pathogenicity and to prioritize candidate therapeutics. We investigate recent hypotheses on a systems biology level to explore mechanistic virus life cycle drivers, and to extract drug repurposing candidates. CoVex renders COVID-19 drug research systems-medicine-ready by giving the scientific community direct access to network medicine algorithms. It is available at https://exbio.wzw.tum.de/covex/.
Collapse
Affiliation(s)
- Sepideh Sadegh
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Julian Matschinske
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - David B Blumenthal
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Gihanna Galindez
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Tim Kacprowski
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Reza Nasirigerdeh
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Mhaned Oubounyt
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Andreas Pichlmair
- Institute of Virology, TUM School of Medicine, Technical University of Munich, München, Germany
| | - Tim Daniel Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Marisol Salgado-Albarrán
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
- Natural Sciences Department, Universidad Autónoma Metropolitana-Cuajimalpa (UAM-C), 05300, Mexico City, Mexico
| | - Julian Späth
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Alexey Stukalov
- Institute of Virology, TUM School of Medicine, Technical University of Munich, München, Germany
| | - Nina K Wenke
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Kevin Yuan
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Josch K Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, München, Germany.
- Computational Biomedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
386
|
Yang Q, Wang Y, Zhang Y, Li F, Xia W, Zhou Y, Qiu Y, Li H, Zhu F. NOREVA: enhanced normalization and evaluation of time-course and multi-class metabolomic data. Nucleic Acids Res 2020; 48:W436-W448. [PMID: 32324219 PMCID: PMC7319444 DOI: 10.1093/nar/gkaa258] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/21/2020] [Accepted: 04/04/2020] [Indexed: 12/23/2022] Open
Abstract
Biological processes (like microbial growth & physiological response) are usually dynamic and require the monitoring of metabolic variation at different time-points. Moreover, there is clear shift from case-control (N=2) study to multi-class (N>2) problem in current metabolomics, which is crucial for revealing the mechanisms underlying certain physiological process, disease metastasis, etc. These time-course and multi-class metabolomics have attracted great attention, and data normalization is essential for removing unwanted biological/experimental variations in these studies. However, no tool (including NOREVA 1.0 focusing only on case-control studies) is available for effectively assessing the performance of normalization method on time-course/multi-class metabolomic data. Thus, NOREVA was updated to version 2.0 by (i) realizing normalization and evaluation of both time-course and multi-class metabolomic data, (ii) integrating 144 normalization methods of a recently proposed combination strategy and (iii) identifying the well-performing methods by comprehensively assessing the largest set of normalizations (168 in total, significantly larger than those 24 in NOREVA 1.0). The significance of this update was extensively validated by case studies on benchmark datasets. All in all, NOREVA 2.0 is distinguished for its capability in identifying well-performing normalization method(s) for time-course and multi-class metabolomics, which makes it an indispensable complement to other available tools. NOREVA can be accessed at https://idrblab.org/noreva/.
Collapse
Affiliation(s)
- Qingxia Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weiqi Xia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation & The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Yunqing Qiu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation & The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Honglin Li
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
387
|
Investigation on the Mechanism of Qubi Formula in Treating Psoriasis Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4683254. [PMID: 32655662 PMCID: PMC7327573 DOI: 10.1155/2020/4683254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/18/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023]
Abstract
Objective To elucidate the pharmacological mechanisms of Qubi Formula (QBF), a traditional Chinese medicine (TCM) formula which has been demonstrated as an effective therapy for psoriasis in China. Methods The Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, BATMAN-TCM database, and literature search were used to excavate the pharmacologically active ingredients of QBF and to predict the potential targets. Psoriasis-related targets were obtained from Therapeutic Target Database (TTD), DrugBank database (DBD), MalaCards database, and DisGeNET database. Then, we established the network concerning the interactions of potential targets of QBF with well-known psoriasis-related targets by using protein-protein interaction (PPI) data in String database. Afterwards, topological parameters (including DNMC, Degree, Closeness, and Betweenness) were calculated to excavate the core targets of Qubi Formula in treating psoriasis (main targets in the PPI network). Cytoscape was used to construct the ingredients-targets core network for Qubi Formula in treating psoriasis, and ClueGO was used to perform GO-BP and KEGG pathway enrichment analysis on these core targets. Results The ingredient-target-disease core network of QBF in treating psoriasis was screened to contain 175 active ingredients, which corresponded to 27 core targets. Additionally, enrichment analysis suggested that targets of QBF in treating psoriasis were mainly clustered into multiple biological processes (associated with nuclear translocation of proteins, cellular response to multiple stimuli (immunoinflammatory factors, oxidative stress, and nutrient substance), lymphocyte activation, regulation of cyclase activity, cell-cell adhesion, and cell death) and related pathways (VEGF, JAK-STAT, TLRs, NF-κB, and lymphocyte differentiation-related pathways), indicating the underlying mechanisms of QBF on psoriasis. Conclusion In this work, we have successfully illuminated that Qubi Formula could relieve a wide variety of pathological factors (such as inflammatory infiltration and abnormal angiogenesis) of psoriasis in a "multicompound, multitarget, and multipathway" manner by using network pharmacology. Moreover, our present outcomes might shed light on the further clinical application of QBF on psoriasis treatment.
Collapse
|
388
|
Machine-Learned Association of Next-Generation Sequencing-Derived Variants in Thermosensitive Ion Channels Genes with Human Thermal Pain Sensitivity Phenotypes. Int J Mol Sci 2020; 21:ijms21124367. [PMID: 32575443 PMCID: PMC7352872 DOI: 10.3390/ijms21124367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/20/2022] Open
Abstract
Genetic association studies have shown their usefulness in assessing the role of ion channels in human thermal pain perception. We used machine learning to construct a complex phenotype from pain thresholds to thermal stimuli and associate it with the genetic information derived from the next-generation sequencing (NGS) of 15 ion channel genes which are involved in thermal perception, including ASIC1, ASIC2, ASIC3, ASIC4, TRPA1, TRPC1, TRPM2, TRPM3, TRPM4, TRPM5, TRPM8, TRPV1, TRPV2, TRPV3, and TRPV4. Phenotypic information was complete in 82 subjects and NGS genotypes were available in 67 subjects. A network of artificial neurons, implemented as emergent self-organizing maps, discovered two clusters characterized by high or low pain thresholds for heat and cold pain. A total of 1071 variants were discovered in the 15 ion channel genes. After feature selection, 80 genetic variants were retained for an association analysis based on machine learning. The measured performance of machine learning-mediated phenotype assignment based on this genetic information resulted in an area under the receiver operating characteristic curve of 77.2%, justifying a phenotype classification based on the genetic information. A further item categorization finally resulted in 38 genetic variants that contributed most to the phenotype assignment. Most of them (10) belonged to the TRPV3 gene, followed by TRPM3 (6). Therefore, the analysis successfully identified the particular importance of TRPV3 and TRPM3 for an average pain phenotype defined by the sensitivity to moderate thermal stimuli.
Collapse
|
389
|
Wang C, Zhao N, Sun K, Zhang Y. A Cancer Gene Module Mining Method Based on Bio-Network of Multi-Omics Gene Groups. Front Oncol 2020; 10:1159. [PMID: 32637361 PMCID: PMC7317001 DOI: 10.3389/fonc.2020.01159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/08/2020] [Indexed: 11/13/2022] Open
Abstract
The initiation, promotion and progression of cancer are highly associated to the environment a human lives in as well as individual genetic factors. In view of the dangers to life and health caused by this abnormally complex systemic disease, many top scientific research institutions around the world have been actively carrying out research in order to discover the pathogenic mechanisms driving cancer occurrence and development. The emergence of high-throughput sequencing technology has greatly advanced oncology research and given rise to the revelation of important oncogenes and the interrelationship among them. Here, we have studied heterogeneous multi-level data within a context of integrated data, and scientifically introduced lncRNA omics data to construct multi-omics bio-network models, allowing the screening of key cancer-related gene groups. We propose a compactness clustering algorithm based on corrected cumulative rank scores, which uses the functional similarity between groups of genes as a distance measure to excavate key gene modules for abnormal regulation contained in gene groups through clustering. We also conducted a survival analysis using our results and found that our model could divide groups of different levels very well. The results also demonstrate that the integration of multi-omics biological data, key gene modules and their dysregulated gene groups can be discovered, which is crucial for cancer research.
Collapse
Affiliation(s)
- Chunyu Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ning Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kai Sun
- Thoracic Surgery Department, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin, China
| | - Ying Zhang
- Department of Pharmacy, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin, China
| |
Collapse
|
390
|
Wang C, Zhang Y, Han S. Its2vec: Fungal Species Identification Using Sequence Embedding and Random Forest Classification. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2468789. [PMID: 32566672 PMCID: PMC7275950 DOI: 10.1155/2020/2468789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/19/2022]
Abstract
Fungi play essential roles in many ecological processes, and taxonomic classification is fundamental for microbial community characterization and vital for the study and preservation of fungal biodiversity. To cope with massive fungal barcode data, tools that can implement extensive volumes of barcode sequences, especially the internal transcribed spacer (ITS) region, are necessary. However, high variation in the ITS region and computational requirements for processing high-dimensional features remain challenging for existing predictors. In this study, we developed Its2vec, a bioinformatics tool for the classification of fungal ITS barcodes to the species level. An ITS database covering more than 25,000 species in a broad range of fungal taxa was assembled. For dimensionality reduction, a word embedding algorithm was used to represent an ITS sequence as a dense low-dimensional vector. A random forest-based classifier was built for species identification. Benchmarking results showed that our model achieved an accuracy comparable to that of several state-of-the-art predictors, and more importantly, it could implement large datasets and greatly reduce dimensionality. We expect the Its2vec model to be helpful for fungal species identification and, thus, for revealing microbial community structures and in deepening our understanding of their functional mechanisms.
Collapse
Affiliation(s)
- Chao Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ying Zhang
- Department of Pharmacy, Heilongjiang Province Land Reclamation Headquarters General Hospital, Harbin 150088, China
| | - Shuguang Han
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 60054, China
| |
Collapse
|
391
|
Yin Z, Wang Q, Yan X, Zhang L, Tang K, Cao Z, Qiu T. Reveal the Regulation Patterns of Prognosis-Related miRNAs and lncRNAs Across Solid Tumors in the Cancer Genome Atlas. Front Cell Dev Biol 2020; 8:368. [PMID: 32523951 PMCID: PMC7261917 DOI: 10.3389/fcell.2020.00368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/24/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The dysregulation of non-coding RNAs (ncRNAs) such as miRNAs and lncRNAs are associated with the pathogenesis and progression in multiple cancers including solid tumors. Comprehensive investigations of prognosis-related ncRNA markers could promote the development of therapeutic strategies for solid tumors, but rarely reported. METHODS By taking advantage of The Cancer Genome Atlas (TCGA), pan-cancer prognosis analysis (PCPA) models were firstly constructed based on miRNA and lncRNA expression profiles of 8,450 samples in 19 solid tumors. Further, the co-occurrence and exclusivity among ncRNA markers were systematically analyzed for different cancers. RESULTS In identified ncRNA makers, 71% of the miRNA markers were shared in multiple cancers, whereas 96% of the lncRNA markers were cancer-specific. Moreover, to analyze the regulation patterns of prognosis-related ncRNAs at the pan-cancer level, miRNA markers were further annotated into eight carcinogenic pathways. Results represented that approximately 86% of these miRNA markers could regulate the PI3K-Akt signaling pathway, while only 48% for the Notch signaling pathway. Finally, among 126 common genes that participated in eight carcinogenic pathways, BCL2, CSNK2A1, EGFR, PDGFRA, and VEGFA were proposed as potential drug targets for multiple cancers. CONCLUSION The prognosis analysis and regulation characteristics of ncRNAs presented in this study may help to facilitate the discovery of anti-cancer drugs for multiple solid tumors.
Collapse
Affiliation(s)
- Zuojing Yin
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qiming Wang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xinmiao Yan
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lu Zhang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Kailin Tang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhiwei Cao
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Tianyi Qiu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
392
|
Wang W, Wang S, Liu T, Ma Y, Huang S, Lei L, Wen A, Ding Y. Resveratrol: Multi-Targets Mechanism on Neurodegenerative Diseases Based on Network Pharmacology. Front Pharmacol 2020; 11:694. [PMID: 32477148 PMCID: PMC7240052 DOI: 10.3389/fphar.2020.00694] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Resveratrol is a natural polyphenol in lots of foods and traditional Chinese medicines, which has shown promising treatment for neurodegenerative diseases (NDs). However, the molecular mechanisms of its action have not been systematically studied yet. In order to elucidate the network pharmacological prospective effects of resveratrol on NDs, we assessed of pharmacokinetics (PK) properties of resveratrol, studied target prediction and network analysis, and discussed interacting pathways using a network pharmacology method. Main PK properties of resveratrol were acquired. A total of 13,612 genes related to NDs, and 138 overlapping genes were determined through matching the 175 potential targets of resveratrol with disease-associated genes. Gene Ontology (GO) function analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were performed to obtain more in-depth understanding of resveratrol on NDs. Accordingly, nodes with high degrees were obtained according using a PPI network, and AKT1, TP53, IL6, CASP3, VEGFA, TNF, MYC, MAPK3, MAPK8, and ALB were identified as hub target genes, which showed better affinity with resveratrol in silico studies. In addition, our experimental results demonstrated that resveratrol markedly enhanced the decreased levels of Bcl-2 and significantly reduced the increased expression of Bax and Caspase-3 in hippocampal neurons induced by glutamate exposure. Western blot results confirmed that resveratrol inhibited glutamate-induced apoptosis of hippocampal neurons partly by regulating the PI3K/AKT/mTOR pathway. In conclusion, we found that resveratrol could target multiple pathways forming a systematic network with pharmacological effects.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Tianlong Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Pharmacy, 940 Hospital of PLA Joint Logistics Support Forces, Lanzhou, China
| | - Yang Ma
- Department of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Shaojie Huang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lu Lei
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
393
|
Qin Z, Xu Q, Hu H, Yu L, Zeng S. Extracellular Vesicles in Renal Cell Carcinoma: Multifaceted Roles and Potential Applications Identified by Experimental and Computational Methods. Front Oncol 2020; 10:724. [PMID: 32457844 PMCID: PMC7221139 DOI: 10.3389/fonc.2020.00724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/16/2020] [Indexed: 12/24/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney cancer. Increasingly evidences indicate that extracellular vesicles (EVs) orchestrate multiple processes in tumorigenesis, metastasis, immune evasion, and drug response of RCC. EVs are lipid membrane-bound vesicles in nanometer size and secreted by almost all cell types into the extracellular milieu. A myriad of bioactive molecules such as RNA, DNA, protein, and lipid are able to be delivered via EVs for the intercellular communication. Hence, the abundant content of EVs is appealing reservoir for biomarker identification through computational analysis and experimental validation. EVs with excellent biocompatibility and biodistribution are natural platforms that can be engineered to offer achievable drug delivery strategies for RCC therapies. Moreover, the multifaceted roles of EVs in RCC progression also provide substantial targets and facilitate EVs-based drug discovery, which will be accelerated by using artificial intelligence approaches. In this review, we summarized the vital roles of EVs in occurrence, metastasis, immune evasion, and drug resistance of RCC. Furthermore, we also recapitulated and prospected the EVs-based potential applications in RCC, including biomarker identification, drug vehicle development as well as drug target discovery.
Collapse
Affiliation(s)
| | | | | | | | - Su Zeng
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang University, Hangzhou, China
| |
Collapse
|
394
|
SSizer: Determining the Sample Sufficiency for Comparative Biological Study. J Mol Biol 2020; 432:3411-3421. [DOI: 10.1016/j.jmb.2020.01.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/26/2019] [Accepted: 01/18/2020] [Indexed: 01/25/2023]
|
395
|
Yan W, Liu X, Wang Y, Han S, Wang F, Liu X, Xiao F, Hu G. Identifying Drug Targets in Pancreatic Ductal Adenocarcinoma Through Machine Learning, Analyzing Biomolecular Networks, and Structural Modeling. Front Pharmacol 2020; 11:534. [PMID: 32425783 PMCID: PMC7204992 DOI: 10.3389/fphar.2020.00534] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death and has an extremely poor prognosis. Thus, identifying new disease-associated genes and targets for PDAC diagnosis and therapy is urgently needed. This requires investigations into the underlying molecular mechanisms of PDAC at both the systems and molecular levels. Herein, we developed a computational method of predicting cancer genes and anticancer drug targets that combined three independent expression microarray datasets of PDAC patients and protein-protein interaction data. First, Support Vector Machine–Recursive Feature Elimination was applied to the gene expression data to rank the differentially expressed genes (DEGs) between PDAC patients and controls. Then, protein-protein interaction networks were constructed based on the DEGs, and a new score comprising gene expression and network topological information was proposed to identify cancer genes. Finally, these genes were validated by “druggability” prediction, survival and common network analysis, and functional enrichment analysis. Furthermore, two integrins were screened to investigate their structures and dynamics as potential drug targets for PDAC. Collectively, 17 disease genes and some stroma-related pathways including extracellular matrix-receptor interactions were predicted to be potential drug targets and important pathways for treating PDAC. The protein-drug interactions and hinge sites predication of ITGAV and ITGA2 suggest potential drug binding residues in the Thigh domain. These findings provide new possibilities for targeted therapeutic interventions in PDAC, which may have further applications in other cancer types.
Collapse
Affiliation(s)
- Wenying Yan
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xingyi Liu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Yibo Wang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Shuqing Han
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Fan Wang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xin Liu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Fei Xiao
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Guang Hu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
396
|
Arroyo MM, Berral-González A, Bueno-Fortes S, Alonso-López D, De Las Rivas J. Mining Drug-Target Associations in Cancer: Analysis of Gene Expression and Drug Activity Correlations. Biomolecules 2020; 10:biom10050667. [PMID: 32344870 PMCID: PMC7277587 DOI: 10.3390/biom10050667] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer is a complex disease affecting millions of people worldwide, with over a hundred clinically approved drugs available. In order to improve therapy, treatment, and response, it is essential to draw better maps of the targets of cancer drugs and possible side interactors. This study presents a large-scale screening method to find associations of cancer drugs with human genes. The analysis is focused on the current collection of Food and Drug Administration (FDA)-approved drugs (which includes about one hundred chemicals). The approach integrates global gene-expression transcriptomic profiles with drug-activity profiles of a set of 60 human cell lines obtained for a collection of chemical compounds (small bioactive molecules). Using a standardized expression for each gene versus standardized activity for each drug, Pearson and Spearman correlations were calculated for all possible pairwise gene-drug combinations. These correlations were used to build a global bipartite network that includes 1007 gene-drug significant associations. The data are integrated into an open web-tool called GEDA (Gene Expression and Drug Activity) which includes a relational view of cancer drugs and genes, disclosing the putative indirect interactions found for FDA-approved drugs as well as the known targets of these drugs. The results also provide insight into the complex action of pharmaceuticals, presenting an alternative view to address predicted pleiotropic effects of the drugs.
Collapse
Affiliation(s)
- Monica M. Arroyo
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain; (A.B.-G.); (S.B.-F.); (D.A.-L.)
- Department of Chemistry, Pontifical Catholic University of Puerto Rico (PCUPR), 00717 Ponce, Puerto Rico
- Correspondence: (M.M.A.); (J.D.L.R.); Tel.: +34-923-294819 (J.D.L.R.)
| | - Alberto Berral-González
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain; (A.B.-G.); (S.B.-F.); (D.A.-L.)
| | - Santiago Bueno-Fortes
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain; (A.B.-G.); (S.B.-F.); (D.A.-L.)
| | - Diego Alonso-López
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain; (A.B.-G.); (S.B.-F.); (D.A.-L.)
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain; (A.B.-G.); (S.B.-F.); (D.A.-L.)
- Correspondence: (M.M.A.); (J.D.L.R.); Tel.: +34-923-294819 (J.D.L.R.)
| |
Collapse
|
397
|
Ciliary Genes in Renal Cystic Diseases. Cells 2020; 9:cells9040907. [PMID: 32276433 PMCID: PMC7226761 DOI: 10.3390/cells9040907] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/27/2020] [Accepted: 04/05/2020] [Indexed: 12/28/2022] Open
Abstract
Cilia are microtubule-based organelles, protruding from the apical cell surface and anchoring to the cytoskeleton. Primary (nonmotile) cilia of the kidney act as mechanosensors of nephron cells, responding to fluid movements by triggering signal transduction. The impaired functioning of primary cilia leads to formation of cysts which in turn contribute to development of diverse renal diseases, including kidney ciliopathies and renal cancer. Here, we review current knowledge on the role of ciliary genes in kidney ciliopathies and renal cell carcinoma (RCC). Special focus is given on the impact of mutations and altered expression of ciliary genes (e.g., encoding polycystins, nephrocystins, Bardet-Biedl syndrome (BBS) proteins, ALS1, Oral-facial-digital syndrome 1 (OFD1) and others) in polycystic kidney disease and nephronophthisis, as well as rare genetic disorders, including syndromes of Joubert, Meckel-Gruber, Bardet-Biedl, Senior-Loken, Alström, Orofaciodigital syndrome type I and cranioectodermal dysplasia. We also show that RCC and classic kidney ciliopathies share commonly disturbed genes affecting cilia function, including VHL (von Hippel-Lindau tumor suppressor), PKD1 (polycystin 1, transient receptor potential channel interacting) and PKD2 (polycystin 2, transient receptor potential cation channel). Finally, we discuss the significance of ciliary genes as diagnostic and prognostic markers, as well as therapeutic targets in ciliopathies and cancer.
Collapse
|
398
|
Meng C, Hu Y, Zhang Y, Guo F. PSBP-SVM: A Machine Learning-Based Computational Identifier for Predicting Polystyrene Binding Peptides. Front Bioeng Biotechnol 2020; 8:245. [PMID: 32296690 PMCID: PMC7137786 DOI: 10.3389/fbioe.2020.00245] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Polystyrene binding peptides (PSBPs) play a key role in the immobilization process. The correct identification of PSBPs is the first step of all related works. In this paper, we proposed a novel support vector machine-based bioinformatic identification model. This model contains four machine learning steps, including feature extraction, feature selection, model training and optimization. In a five-fold cross validation test, this model achieves 90.38, 84.62, 87.50, and 0.90% SN, SP, ACC, and AUC, respectively. The performance of this model outperforms the state-of-the-art identifier in terms of the SN and ACC with a smaller feature set. Furthermore, we constructed a web server that includes the proposed model, which is freely accessible at http://server.malab.cn/PSBP-SVM/index.jsp.
Collapse
Affiliation(s)
- Chaolu Meng
- College of Intelligence and Computing, Tianjin University, Tianjin, China.,College of Computer and Information Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Yang Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, China
| | - Ying Zhang
- Department of Pharmacy, Heilongjiang Province Land Reclamation Headquarters General Hospital, Harbin, China
| | - Fei Guo
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| |
Collapse
|
399
|
Bagherian M, Kim RB, Jiang C, Sartor MA, Derksen H, Najarian K. Coupled matrix-matrix and coupled tensor-matrix completion methods for predicting drug-target interactions. Brief Bioinform 2020; 22:2161-2171. [PMID: 32186716 PMCID: PMC7986629 DOI: 10.1093/bib/bbaa025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/27/2020] [Accepted: 02/17/2020] [Indexed: 12/20/2022] Open
Abstract
Predicting the interactions between drugs and targets plays an important role in the process of new drug discovery, drug repurposing (also known as drug repositioning). There is a need to develop novel and efficient prediction approaches in order to avoid the costly and laborious process of determining drug–target interactions (DTIs) based on experiments alone. These computational prediction approaches should be capable of identifying the potential DTIs in a timely manner. Matrix factorization methods have been proven to be the most reliable group of methods. Here, we first propose a matrix factorization-based method termed ‘Coupled Matrix–Matrix Completion’ (CMMC). Next, in order to utilize more comprehensive information provided in different databases and incorporate multiple types of scores for drug–drug similarities and target–target relationship, we then extend CMMC to ‘Coupled Tensor–Matrix Completion’ (CTMC) by considering drug–drug and target–target similarity/interaction tensors. Results: Evaluation on two benchmark datasets, DrugBank and TTD, shows that CTMC outperforms the matrix-factorization-based methods: GRMF, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$L_{2,1}$\end{document}-GRMF, NRLMF and NRLMF\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$\beta $\end{document}. Based on the evaluation, CMMC and CTMC outperform the above three methods in term of area under the curve, F1 score, sensitivity and specificity in a considerably shorter run time.
Collapse
Affiliation(s)
- Maryam Bagherian
- Corresponding author: Maryam Bagherian, Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 48109, USA.
| | | | | | | | | | | |
Collapse
|
400
|
Prediction of Side Effects Using Comprehensive Similarity Measures. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1357630. [PMID: 32190647 PMCID: PMC7064827 DOI: 10.1155/2020/1357630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/19/2020] [Accepted: 02/04/2020] [Indexed: 11/17/2022]
Abstract
Identifying the potential side effects of drugs is crucial in clinical trials in the pharmaceutical industry. The existing side effect prediction methods mainly focus on the chemical and biological properties of drugs. This study proposes a method that uses diverse information such as drug-drug interactions from DrugBank, drug-drug interactions from network, single nucleotide polymorphisms, and side effect anatomical hierarchy as well as chemical structures, indications, and targets. The proposed method is based on the assumption that properties used in drug repositioning studies could be utilized to predict side effects because the phenotypic expression of a side effect is similar to that of the disease. The prediction results using the proposed method showed a 3.5% improvement in the area under the curve (AUC) over that obtained when only chemical, indication, and target features were used. The random forest model delivered outstanding results for all combinations of feature types. Finally, after identifying candidate side effects of drugs using the proposed method, the following four popular drugs were discussed: (1) dasatinib, (2) sitagliptin, (3) vorinostat, and (4) clonidine.
Collapse
|