1
|
Nguyen-Vo TH, Do TTT, Nguyen BP. Multitask Learning on Graph Convolutional Residual Neural Networks for Screening of Multitarget Anticancer Compounds. J Chem Inf Model 2024. [PMID: 39197175 DOI: 10.1021/acs.jcim.4c00643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Recently, various modern experimental screening pipelines and assays have been developed to find promising anticancer drug candidates. However, it is time-consuming and almost infeasible to screen an immense number of compounds for anticancer activity via experimental approaches. To partially address this issue, several computational advances have been proposed. In this study, we present iACP-GCR, a model based on multitask learning on graph convolutional residual neural networks with two types of shortcut connections, to identify multitarget anticancer compounds. In our architecture, the graph convolutional residual neural networks are shared by all the prediction tasks before being separately customized. The NCI-60 data set, one of the most reliable and well-known sources of experimentally verified compounds, was used to develop our model. From that data set, we collected and refined data about compounds screened across nine cancer types (panels), including breast, central nervous system, colon, leukemia, nonsmall cell lung, melanoma, ovarian, prostate, and renal, for model training and evaluation. The model performance evaluated on an independent test set shows that iACP-GCR surpasses the three advanced computational methods for multitask learning. The integration of two shortcut connection types in the shared networks also improves the prediction efficiency. We also deployed the model as a public web server to assist the research community in screening potential anticancer compounds.
Collapse
Affiliation(s)
- Thanh-Hoang Nguyen-Vo
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Trang T T Do
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Binh P Nguyen
- Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand
| |
Collapse
|
2
|
Wang J, Hong M, Cheng Y, Wang X, Li D, Chen G, Bao B, Song J, Du X, Yang C, Zheng L, Tong Q. Targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for malate-aspartate shuttle and tumour progression. Clin Transl Med 2024; 14:e1680. [PMID: 38769668 PMCID: PMC11106511 DOI: 10.1002/ctm2.1680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/28/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND A series of studies have demonstrated the emerging involvement of transfer RNA (tRNA) processing during the progression of tumours. Nevertheless, the roles and regulating mechanisms of tRNA processing genes in neuroblastoma (NB), the prevalent malignant tumour outside the brain in children, are yet unknown. METHODS Analysis of multi-omics results was conducted to identify crucial regulators of downstream tRNA processing genes. Co-immunoprecipitation and mass spectrometry methods were utilised to measure interaction between proteins. The impact of transcriptional regulators on expression of downstream genes was measured by dual-luciferase reporter, chromatin immunoprecipitation, western blotting and real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR) methods. Studies have been conducted to reveal impact and mechanisms of transcriptional regulators on biological processes of NB. Survival differences were analysed using the log-rank test. RESULTS c-Myc was identified as a transcription factor driving tRNA processing gene expression and subsequent malate-aspartate shuttle (MAS) in NB cells. Mechanistically, c-Myc directly promoted the expression of glutamyl-prolyl-tRNA synthetase (EPRS) and leucyl-tRNA synthetase (LARS), resulting in translational up-regulation of glutamic-oxaloacetic transaminase 1 (GOT1) as well as malate dehydrogenase 1 (MDH1) via inhibiting general control nonrepressed 2 or activating mechanistic target of rapamycin signalling. Meanwhile, lamin A (LMNA) inhibited c-Myc transactivation via physical interaction, leading to suppression of MAS, aerobic glycolysis, tumourigenesis and aggressiveness. Pre-clinically, lobeline was discovered as a LMNA-binding compound to facilitate its interaction with c-Myc, which inhibited aminoacyl-tRNA synthetase expression, MAS and tumour progression of NB, as well as growth of organoid derived from c-Myc knock-in mice. Low levels of LMNA or elevated expression of c-Myc, EPRS, LARS, GOT1 or MDH1 were linked to a worse outcome and a shorter survival time of clinical NB patients. CONCLUSIONS These results suggest that targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for MAS and tumour progression.
Collapse
Affiliation(s)
- Jianqun Wang
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Mei Hong
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Yang Cheng
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Xiaojing Wang
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
- Department of GeriatricsUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Dan Li
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Guo Chen
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Banghe Bao
- Department of PathologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Jiyu Song
- Department of PathologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Xinyi Du
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Chunhui Yang
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Liduan Zheng
- Department of PathologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| | - Qiangsong Tong
- Department of Pediatric SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceP. R. China
| |
Collapse
|
3
|
Son J, Kim D. Applying network link prediction in drug discovery: an overview of the literature. Expert Opin Drug Discov 2024; 19:43-56. [PMID: 37794688 DOI: 10.1080/17460441.2023.2267020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/02/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION Network representation can give a holistic view of relationships for biomedical entities through network topology. Link prediction estimates the probability of link formation between the pair of unconnected nodes. In the drug discovery process, the link prediction method not only enables the detection of connectivity patterns but also predicts the effects of one biomedical entity to multiple entities simultaneously and vice versa, which is useful for many applications. AREAS COVERED The authors provide a comprehensive overview of network link prediction in drug discovery. Link prediction methodologies such as similarity-based approaches, embedding-based approaches, probabilistic model-based approaches, and preprocessing methods are summarized with examples. In addition to describing their properties and limitations, the authors discuss the applications of link prediction in drug discovery based on the relationship between biomedical concepts. EXPERT OPINION Link prediction is a powerful method to infer the existence of novel relationships in drug discovery. However, link prediction has been hampered by the sparsity of data and the lack of negative links in biomedical networks. With preprocessing to balance positive and negative samples and the collection of more data, the authors believe it is possible to develop more reliable link prediction methods that can become invaluable tools for successful drug discovery.
Collapse
Affiliation(s)
- Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
4
|
Huang Y, Huang HY, Chen Y, Lin YCD, Yao L, Lin T, Leng J, Chang Y, Zhang Y, Zhu Z, Ma K, Cheng YN, Lee TY, Huang HD. A Robust Drug-Target Interaction Prediction Framework with Capsule Network and Transfer Learning. Int J Mol Sci 2023; 24:14061. [PMID: 37762364 PMCID: PMC10531393 DOI: 10.3390/ijms241814061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Drug-target interactions (DTIs) are considered a crucial component of drug design and drug discovery. To date, many computational methods were developed for drug-target interactions, but they are insufficiently informative for accurately predicting DTIs due to the lack of experimentally verified negative datasets, inaccurate molecular feature representation, and ineffective DTI classifiers. Therefore, we address the limitations of randomly selecting negative DTI data from unknown drug-target pairs by establishing two experimentally validated datasets and propose a capsule network-based framework called CapBM-DTI to capture hierarchical relationships of drugs and targets, which adopts pre-trained bidirectional encoder representations from transformers (BERT) for contextual sequence feature extraction from target proteins through transfer learning and the message-passing neural network (MPNN) for the 2-D graph feature extraction of compounds to accurately and robustly identify drug-target interactions. We compared the performance of CapBM-DTI with state-of-the-art methods using four experimentally validated DTI datasets of different sizes, including human (Homo sapiens) and worm (Caenorhabditis elegans) species datasets, as well as three subsets (new compounds, new proteins, and new pairs). Our results demonstrate that the proposed model achieved robust performance and powerful generalization ability in all experiments. The case study on treating COVID-19 demonstrates the applicability of the model in virtual screening.
Collapse
Affiliation(s)
- Yixian Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yigang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Lantian Yao
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Tianxiu Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Junlin Leng
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yuan Chang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yuntian Zhang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Zihao Zhu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Kun Ma
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| | - Yeong-Nan Cheng
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; (Y.-N.C.)
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; (Y.-N.C.)
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (Y.H.); (Y.C.); (J.L.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen 518172, China; (L.Y.); (Y.C.)
| |
Collapse
|
5
|
Chen ZH, Zhao BW, Li JQ, Guo ZH, You ZH. GraphCPIs: A novel graph-based computational model for potential compound-protein interactions. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:721-728. [PMID: 37251691 PMCID: PMC10209012 DOI: 10.1016/j.omtn.2023.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/28/2023] [Indexed: 05/31/2023]
Abstract
Identifying proteins that interact with drug compounds has been recognized as an important part in the process of drug discovery. Despite extensive efforts that have been invested in predicting compound-protein interactions (CPIs), existing traditional methods still face several challenges. The computer-aided methods can identify high-quality CPI candidates instantaneously. In this research, a novel model is named GraphCPIs, proposed to improve the CPI prediction accuracy. First, we establish the adjacent matrix of entities connected to both drugs and proteins from the collected dataset. Then, the feature representation of nodes could be obtained by using the graph convolutional network and Grarep embedding model. Finally, an extreme gradient boosting (XGBoost) classifier is exploited to identify potential CPIs based on the stacked two kinds of features. The results demonstrate that GraphCPIs achieves the best performance, whose average predictive accuracy rate reaches 90.09%, average area under the receiver operating characteristic curve is 0.9572, and the average area under the precision and recall curve is 0.9621. Moreover, comparative experiments reveal that our method surpasses the state-of-the-art approaches in the field of accuracy and other indicators with the same experimental environment. We believe that the GraphCPIs model will provide valuable insight to discover novel candidate drug-related proteins.
Collapse
Affiliation(s)
- Zhan-Heng Chen
- Department of Clinical Anesthesiology, Faculty of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Bo-Wei Zhao
- The Xinjiang Technical Institute of Physics & Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Jian-Qiang Li
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhen-Hao Guo
- Institute of Machine Learning and Systems Biology, School of Electronics and Information Engineering, Tongji University, Caoan Road 4800, Shanghai 201804, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710129, China
| |
Collapse
|
6
|
Majd H, Amin S, Ghazizadeh Z, Cesiulis A, Arroyo E, Lankford K, Majd A, Farahvashi S, Chemel AK, Okoye M, Scantlen MD, Tchieu J, Calder EL, Le Rouzic V, Shibata B, Arab A, Goodarzi H, Pasternak G, Kocsis JD, Chen S, Studer L, Fattahi F. Deriving Schwann cells from hPSCs enables disease modeling and drug discovery for diabetic peripheral neuropathy. Cell Stem Cell 2023; 30:632-647.e10. [PMID: 37146583 PMCID: PMC10249419 DOI: 10.1016/j.stem.2023.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 01/11/2023] [Accepted: 04/10/2023] [Indexed: 05/07/2023]
Abstract
Schwann cells (SCs) are the primary glia of the peripheral nervous system. SCs are involved in many debilitating disorders, including diabetic peripheral neuropathy (DPN). Here, we present a strategy for deriving SCs from human pluripotent stem cells (hPSCs) that enables comprehensive studies of SC development, physiology, and disease. hPSC-derived SCs recapitulate the molecular features of primary SCs and are capable of in vitro and in vivo myelination. We established a model of DPN that revealed the selective vulnerability of SCs to high glucose. We performed a high-throughput screen and found that an antidepressant drug, bupropion, counteracts glucotoxicity in SCs. Treatment of hyperglycemic mice with bupropion prevents their sensory dysfunction, SC death, and myelin damage. Further, our retrospective analysis of health records revealed that bupropion treatment is associated with a lower incidence of neuropathy among diabetic patients. These results highlight the power of this approach for identifying therapeutic candidates for DPN.
Collapse
Affiliation(s)
- Homa Majd
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Sadaf Amin
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zaniar Ghazizadeh
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrius Cesiulis
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Edgardo Arroyo
- Neuroscience Research Center, Yale University School of Medicine and VA Healthcare System, West Haven, CT 06516, USA; Department of Neurology, Yale University School of Medicine and VA Healthcare System, West Haven, CT 06516, USA
| | - Karen Lankford
- Neuroscience Research Center, Yale University School of Medicine and VA Healthcare System, West Haven, CT 06516, USA; Department of Neurology, Yale University School of Medicine and VA Healthcare System, West Haven, CT 06516, USA
| | - Alireza Majd
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Sina Farahvashi
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Angeline K Chemel
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Mesomachukwu Okoye
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Megan D Scantlen
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA
| | - Jason Tchieu
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Elizabeth L Calder
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Valerie Le Rouzic
- Molecular Pharmacology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Neurology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Bradley Shibata
- Biological Electron Microscopy Facility, UCD, Davis, CA 95616, USA
| | - Abolfazl Arab
- Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94158, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94158, USA; Department of Urology, UCSF, San Francisco, CA 94158, USA
| | - Gavril Pasternak
- Molecular Pharmacology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Neurology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Jeffery D Kocsis
- Neuroscience Research Center, Yale University School of Medicine and VA Healthcare System, West Haven, CT 06516, USA; Department of Neurology, Yale University School of Medicine and VA Healthcare System, West Haven, CT 06516, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA; Center of Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA.
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94158, USA; Program in Craniofacial Biology, UCSF, San Francisco, CA 94110, USA.
| |
Collapse
|
7
|
Hoang VT, Jeon HJ, You ES, Yoon Y, Jung S, Lee OJ. Graph Representation Learning and Its Applications: A Survey. SENSORS (BASEL, SWITZERLAND) 2023; 23:4168. [PMID: 37112507 PMCID: PMC10144941 DOI: 10.3390/s23084168] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 06/19/2023]
Abstract
Graphs are data structures that effectively represent relational data in the real world. Graph representation learning is a significant task since it could facilitate various downstream tasks, such as node classification, link prediction, etc. Graph representation learning aims to map graph entities to low-dimensional vectors while preserving graph structure and entity relationships. Over the decades, many models have been proposed for graph representation learning. This paper aims to show a comprehensive picture of graph representation learning models, including traditional and state-of-the-art models on various graphs in different geometric spaces. First, we begin with five types of graph embedding models: graph kernels, matrix factorization models, shallow models, deep-learning models, and non-Euclidean models. In addition, we also discuss graph transformer models and Gaussian embedding models. Second, we present practical applications of graph embedding models, from constructing graphs for specific domains to applying models to solve tasks. Finally, we discuss challenges for existing models and future research directions in detail. As a result, this paper provides a structured overview of the diversity of graph embedding models.
Collapse
Affiliation(s)
- Van Thuy Hoang
- Department of Artificial Intelligence, The Catholic University of Korea, 43, Jibong-ro, Bucheon-si 14662, Gyeonggi-do, Republic of Korea; (V.T.H.); (E.-S.Y.)
| | - Hyeon-Ju Jeon
- Data Assimilation Group, Korea Institute of Atmospheric Prediction Systems (KIAPS), 35, Boramae-ro 5-gil, Dongjak-gu, Seoul 07071, Republic of Korea;
| | - Eun-Soon You
- Department of Artificial Intelligence, The Catholic University of Korea, 43, Jibong-ro, Bucheon-si 14662, Gyeonggi-do, Republic of Korea; (V.T.H.); (E.-S.Y.)
| | - Yoewon Yoon
- Department of Social Welfare, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 04620, Republic of Korea;
| | - Sungyeop Jung
- Semiconductor Devices and Circuits Laboratory, Advanced Institute of Convergence Technology (AICT), Seoul National University, 145, Gwanggyo-ro, Yeongtong-gu, Suwon-si 16229, Gyeonggi-do, Republic of Korea;
| | - O-Joun Lee
- Department of Artificial Intelligence, The Catholic University of Korea, 43, Jibong-ro, Bucheon-si 14662, Gyeonggi-do, Republic of Korea; (V.T.H.); (E.-S.Y.)
| |
Collapse
|
8
|
Shokri Garjan H, Omidi Y, Poursheikhali Asghari M, Ferdousi R. In-silico computational approaches to study microbiota impacts on diseases and pharmacotherapy. Gut Pathog 2023; 15:10. [PMID: 36882861 PMCID: PMC9990230 DOI: 10.1186/s13099-023-00535-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/21/2023] [Indexed: 03/09/2023] Open
Abstract
Microorganisms have been linked to a variety of critical human disease, thanks to advances in sequencing technology and microbiology. The growing recognition of human microbe-disease relationships provides crucial insights into the underlying disease process from the perspective of pathogens, which is extremely useful for pathogenesis research, early diagnosis, and precision medicine and therapy. Microbe-based analysis in terms of diseases and related drug discovery can predict new connections/mechanisms and provide new concepts. These phenomena have been studied via various in-silico computational approaches. This review aims to elaborate on the computational works conducted on the microbe-disease and microbe-drug topics, discuss the computational model approaches used for predicting associations and provide comprehensive information on the related databases. Finally, we discussed potential prospects and obstacles in this field of study, while also outlining some recommendations for further enhancing predictive capabilities.
Collapse
Affiliation(s)
- Hassan Shokri Garjan
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, Fort Lauderdale, FL, USA
| | | | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Shi C, Nie F, Hu Y, Xu Y, Chen L, Ma X, Luo Q. MedChemLens: An Interactive Visual Tool to Support Direction Selection in Interdisciplinary Experimental Research of Medicinal Chemistry. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2023; 29:63-73. [PMID: 36166547 DOI: 10.1109/tvcg.2022.3209434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Interdisciplinary experimental science (e.g., medicinal chemistry) refers to the disciplines that integrate knowledge from different scientific backgrounds and involve experiments in the research process. Deciding "in what direction to proceed" is critical for the success of the research in such disciplines, since the time, money, and resource costs of the subsequent research steps depend largely on this decision. However, such a direction identification task is challenging in that researchers need to integrate information from large-scale, heterogeneous materials from all associated disciplines and summarize the related publications of which the core contributions are often showcased in diverse formats. The task also requires researchers to estimate the feasibility and potential in future experiments in the selected directions. In this work, we selected medicinal chemistry as a case and presented an interactive visual tool, MedChemLens, to assist medicinal chemists in choosing their intended directions of research. This task is also known as drug target (i.e., disease-linked proteins) selection. Given a candidate target name, MedChemLens automatically extracts the molecular features of drug compounds from chemical papers and clinical trial records, organizes them based on the drug structures, and interactively visualizes factors concerning subsequent experiments. We evaluated MedChemLens through a within-subjects study (N=16). Compared with the control condition (i.e., unrestricted online search without using our tool), participants who only used MedChemLens reported faster search, better-informed selections, higher confidence in their selections, and lower cognitive load.
Collapse
|
10
|
Hermawan A, Wulandari F, Hanif N, Utomo RY, Jenie RI, Ikawati M, Tafrihani AS. Identification of potential targets of the curcumin analog CCA-1.1 for glioblastoma treatment : integrated computational analysis and in vitro study. Sci Rep 2022; 12:13928. [PMID: 35977996 PMCID: PMC9385707 DOI: 10.1038/s41598-022-18348-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 08/10/2022] [Indexed: 11/09/2022] Open
Abstract
The treatment of glioblastoma multiforme (GBM) is challenging owing to its localization in the brain, the limited capacity of brain cells to repair, resistance to conventional therapy, and its aggressiveness. Curcumin has anticancer activity against aggressive cancers, such as leukemia, and GBM; however, its application is limited by its low solubility and bioavailability. Chemoprevention curcumin analog 1.1 (CCA-1.1), a curcumin analog, has better solubility and stability than those of curcumin. In this study, we explored potential targets of CCA-1.1 in GBM (PTCGs) by an integrated computational analysis and in vitro study. Predicted targets of CCA-1.1 obtained using various databases were subjected to comprehensive downstream analyses, including functional annotation, disease and drug association analyses, protein–protein interaction network analyses, analyses of genetic alterations, expression, and associations with survival and immune cell infiltration. Our integrative bioinformatics analysis revealed four candidate targets of CCA-1.1 in GBM: TP53, EGFR, AKT1, and CASP3. In addition to targeting specific proteins with regulatory effects in GBM, CCA-1.1 has the capacity to modulate the immunological milieu. Cytotoxicity of CCA-1.1 was lower than TMZ with an IC50 value of 9.8 μM compared to TMZ with an IC50 of 40 μM. mRNA sequencing revealed EGFR transcript variant 8 was upregulated, whereas EGFRvIII was downregulated in U87 cells after treatment with CCA-1.1. Furthermore, a molecular docking analysis suggested that CCA-1.1 inhibits EGFR with various mutations in GBM, which was confirmed using molecular dynamics simulation, wherein the binding between CCA-1.1 with the mutant EGFR L861Q was stable. For successful clinical translation, the effects of CCA-1.1 need to be confirmed in laboratory studies and clinical trials.
Collapse
Affiliation(s)
- Adam Hermawan
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia. .,Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia.
| | - Febri Wulandari
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| | - Naufa Hanif
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| | - Rohmad Yudi Utomo
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia.,Laboratory of Medicinal Chemistry, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| | - Riris Istighfari Jenie
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia.,Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| | - Muthi Ikawati
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia.,Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| | - Ahmad Syauqy Tafrihani
- Faculty of Pharmacy, Cancer Chemoprevention Research Center, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| |
Collapse
|
11
|
Sánchez-Ruiz A, Colmenarejo G. Systematic Analysis and Prediction of the Target Space of Bioactive Food Compounds: Filling the Chemobiological Gaps. J Chem Inf Model 2022; 62:3734-3751. [PMID: 35938782 DOI: 10.1021/acs.jcim.2c00888] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Food compounds and their molecular interactions are crucial for health and provide new chemotypes and targets for drug and nutraceutic design. Here, we retrieve and analyze the complete set of published interactions of food compounds with human proteins using the FooDB as a compound set and ChEMBL as a source of interactions. The data are analyzed in terms of 19 target classes and 19 compound classes, showing a small fraction of target assignment for the compounds (1.6%) and unraveling multiple gaps in the chemobiological space for these molecules. By using well-established cheminformatic approaches [similarity ensemble approach (SEA) combined with the maximum Tanimoto coefficient to the nearest bioactive, "SEA + TC"], we achieve a much enhanced target assignment (64.2%), filling many of the gaps with target hypothesis for fast focused testing. By publishing these data sets and analyses, we expect to provide a set of resources to speed up the full clarification of the chemobiological space of food compounds, opening new opportunities for drug and nutraceutic design.
Collapse
Affiliation(s)
- Andrés Sánchez-Ruiz
- Biostatistics and Bioinformatics Unit, IMDEA Food, CEI UAM+CSIC, E28049 Madrid, Spain
| | - Gonzalo Colmenarejo
- Biostatistics and Bioinformatics Unit, IMDEA Food, CEI UAM+CSIC, E28049 Madrid, Spain
| |
Collapse
|
12
|
Zheng J, Xiao X, Qiu WR. DTI-BERT: Identifying Drug-Target Interactions in Cellular Networking Based on BERT and Deep Learning Method. Front Genet 2022; 13:859188. [PMID: 35754843 PMCID: PMC9213727 DOI: 10.3389/fgene.2022.859188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Drug–target interactions (DTIs) are regarded as an essential part of genomic drug discovery, and computational prediction of DTIs can accelerate to find the lead drug for the target, which can make up for the lack of time-consuming and expensive wet-lab techniques. Currently, many computational methods predict DTIs based on sequential composition or physicochemical properties of drug and target, but further efforts are needed to improve them. In this article, we proposed a new sequence-based method for accurately identifying DTIs. For target protein, we explore using pre-trained Bidirectional Encoder Representations from Transformers (BERT) to extract sequence features, which can provide unique and valuable pattern information. For drug molecules, Discrete Wavelet Transform (DWT) is employed to generate information from drug molecular fingerprints. Then we concatenate the feature vectors of the DTIs, and input them into a feature extraction module consisting of a batch-norm layer, rectified linear activation layer and linear layer, called BRL block and a Convolutional Neural Networks module to extract DTIs features further. Subsequently, a BRL block is used as the prediction engine. After optimizing the model based on contrastive loss and cross-entropy loss, it gave prediction accuracies of the target families of G Protein-coupled receptors, ion channels, enzymes, and nuclear receptors up to 90.1, 94.7, 94.9, and 89%, which indicated that the proposed method can outperform the existing predictors. To make it as convenient as possible for researchers, the web server for the new predictor is freely accessible at: https://bioinfo.jcu.edu.cn/dtibert or http://121.36.221.79/dtibert/. The proposed method may also be a potential option for other DITs.
Collapse
Affiliation(s)
- Jie Zheng
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Xuan Xiao
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Wang-Ren Qiu
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| |
Collapse
|
13
|
Novitasari D, Jenie RI, Kato JY, Meiyanto E. The integrative bioinformatic analysis deciphers the predicted molecular target gene and pathway from curcumin derivative CCA-1.1 against triple-negative breast cancer (TNBC). J Egypt Natl Canc Inst 2021; 33:19. [PMID: 34337682 DOI: 10.1186/s43046-021-00077-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/16/2021] [Indexed: 03/20/2023] Open
Abstract
BACKGROUND The poor outcomes from triple-negative breast cancer (TNBC) therapy are mainly because of TNBC cells' heterogeneity, and chemotherapy is the current approach in TNBC treatment. A previous study reported that CCA-1.1, the alcohol-derivative from monocarbonyl PGV-1, exhibits anticancer activities against several cancer cells, as well as in TNBC. This time, we utilized an integrative bioinformatics approach to identify potential biomarkers and molecular mechanisms of CCA-1.1 in inhibiting proliferation in TNBC cells. METHODS Genomics data expression were collected through UALCAN, derived initially from TCGA-BRCA data, and selected for TNBC-only cases. We predict CCA-1.1 potential targets using SMILES-based similarity functions across six public web tools (BindingDB, DINIES, Swiss Target Prediction, Polypharmacology browser/PPB, Similarity Ensemble Approach/SEA, and TargetNet). The overlapping genes between the CCA-1.1 target and TNBC (CPTGs) were selected and used in further assessment. Gene ontology (GO) enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) network analysis were generated in WebGestalt. The protein-protein interaction (PPI) network was established in STRING-DB, and then the hub-genes were defined through Cytoscape. The hub-gene's survival analysis was processed via CTGS web tools using TCGA database. RESULTS KEGG pathway analysis pointed to cell cycle process which enriched in CCA-1.1 potential targets. We also identified nine CPTGs that are responsible in mitosis, including AURKB, PLK1, CDK1, TPX2, AURKA, KIF11, CDC7, CHEK1, and CDC25B. CONCLUSION We suggested CCA-1.1 possibly regulated cell cycle process during mitosis, which led to cell death. These findings needed to be investigated through experimental studies to reinforce scientific data of CCA-1.1 therapy against TNBC.
Collapse
Affiliation(s)
- Dhania Novitasari
- Doctoral Student in the Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.,Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Riris Istighfari Jenie
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.,Macromolecular Engineering Laboratory, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Jun-Ya Kato
- Laboratory of Tumor Cell Biology, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Edy Meiyanto
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia. .,Macromolecular Engineering Laboratory, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta, 55281, Indonesia.
| |
Collapse
|
14
|
Fathima S, Sinha S, Donakonda S. Network Analysis Identifies Drug Targets and Small Molecules to Modulate Apoptosis Resistant Cancers. Cancers (Basel) 2021; 13:851. [PMID: 33670487 PMCID: PMC7922238 DOI: 10.3390/cancers13040851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/20/2022] Open
Abstract
Programed cell death or apoptosis fails to induce cell death in many recalcitrant cancers. Thus, there is an emerging need to activate the alternate cell death pathways in such cancers. In this study, we analyzed the apoptosis-resistant colon adenocarcinoma, glioblastoma multiforme, and small cell lung cancers transcriptome profiles. We extracted clusters of non-apoptotic cell death genes from each cancer to understand functional networks affected by these genes and their role in the induction of cell death when apoptosis fails. We identified transcription factors regulating cell death genes and protein-protein interaction networks to understand their role in regulating cell death mechanisms. Topological analysis of networks yielded FANCD2 (ferroptosis, negative regulator, down), NCOA4 (ferroptosis, up), IKBKB (alkaliptosis, down), and RHOA (entotic cell death, down) as potential drug targets in colon adenocarcinoma, glioblastoma multiforme, small cell lung cancer phenotypes respectively. We also assessed the miRNA association with the drug targets. We identified tumor growth-related interacting partners based on the pathway information of drug-target interaction networks. The protein-protein interaction binding site between the drug targets and their interacting proteins provided an opportunity to identify small molecules that can modulate the activity of functional cell death interactions in each cancer. Overall, our systematic screening of non-apoptotic cell death-related genes uncovered targets helpful for cancer therapy.
Collapse
Affiliation(s)
- Samreen Fathima
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, MS Ramaiah University of Applied Sciences, Bengaluru 560054, India;
| | - Swati Sinha
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, MS Ramaiah University of Applied Sciences, Bengaluru 560054, India;
| | - Sainitin Donakonda
- School of Medicine, Institute of Molecular Immunology and Experimental Oncology, Klinikum Rechts Der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
15
|
Makarov I, Kiselev D, Nikitinsky N, Subelj L. Survey on graph embeddings and their applications to machine learning problems on graphs. PeerJ Comput Sci 2021; 7:e357. [PMID: 33817007 PMCID: PMC7959646 DOI: 10.7717/peerj-cs.357] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/18/2020] [Indexed: 05/13/2023]
Abstract
Dealing with relational data always required significant computational resources, domain expertise and task-dependent feature engineering to incorporate structural information into a predictive model. Nowadays, a family of automated graph feature engineering techniques has been proposed in different streams of literature. So-called graph embeddings provide a powerful tool to construct vectorized feature spaces for graphs and their components, such as nodes, edges and subgraphs under preserving inner graph properties. Using the constructed feature spaces, many machine learning problems on graphs can be solved via standard frameworks suitable for vectorized feature representation. Our survey aims to describe the core concepts of graph embeddings and provide several taxonomies for their description. First, we start with the methodological approach and extract three types of graph embedding models based on matrix factorization, random-walks and deep learning approaches. Next, we describe how different types of networks impact the ability of models to incorporate structural and attributed data into a unified embedding. Going further, we perform a thorough evaluation of graph embedding applications to machine learning problems on graphs, among which are node classification, link prediction, clustering, visualization, compression, and a family of the whole graph embedding algorithms suitable for graph classification, similarity and alignment problems. Finally, we overview the existing applications of graph embeddings to computer science domains, formulate open problems and provide experiment results, explaining how different networks properties result in graph embeddings quality in the four classic machine learning problems on graphs, such as node classification, link prediction, clustering and graph visualization. As a result, our survey covers a new rapidly growing field of network feature engineering, presents an in-depth analysis of models based on network types, and overviews a wide range of applications to machine learning problems on graphs.
Collapse
Affiliation(s)
- Ilya Makarov
- HSE University, Moscow, Russia
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | | | - Nikita Nikitinsky
- Big Data Research Center, National University of Science and Technology MISIS, Moscow, Russia
| | - Lovro Subelj
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
16
|
Wang C, Kurgan L. Survey of Similarity-Based Prediction of Drug-Protein Interactions. Curr Med Chem 2021; 27:5856-5886. [PMID: 31393241 DOI: 10.2174/0929867326666190808154841] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/16/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022]
Abstract
Therapeutic activity of a significant majority of drugs is determined by their interactions with proteins. Databases of drug-protein interactions (DPIs) primarily focus on the therapeutic protein targets while the knowledge of the off-targets is fragmented and partial. One way to bridge this knowledge gap is to employ computational methods to predict protein targets for a given drug molecule, or interacting drugs for given protein targets. We survey a comprehensive set of 35 methods that were published in high-impact venues and that predict DPIs based on similarity between drugs and similarity between protein targets. We analyze the internal databases of known PDIs that these methods utilize to compute similarities, and investigate how they are linked to the 12 publicly available source databases. We discuss contents, impact and relationships between these internal and source databases, and well as the timeline of their releases and publications. The 35 predictors exploit and often combine three types of similarities that consider drug structures, drug profiles, and target sequences. We review the predictive architectures of these methods, their impact, and we explain how their internal DPIs databases are linked to the source databases. We also include a detailed timeline of the development of these predictors and discuss the underlying limitations of the current resources and predictive tools. Finally, we provide several recommendations concerning the future development of the related databases and methods.
Collapse
Affiliation(s)
- Chen Wang
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, United States
| |
Collapse
|
17
|
Li H, Pei F, Taylor DL, Bahar I. QuartataWeb: Integrated Chemical-Protein-Pathway Mapping for Polypharmacology and Chemogenomics. Bioinformatics 2020; 36:3935-3937. [PMID: 32221612 PMCID: PMC7320630 DOI: 10.1093/bioinformatics/btaa210] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/04/2020] [Accepted: 03/24/2020] [Indexed: 01/31/2023] Open
Abstract
SUMMARY QuartataWeb is a user-friendly server developed for polypharmacological and chemogenomics analyses. Users can easily obtain information on experimentally verified (known) and computationally predicted (new) interactions between 5494 drugs and 2807 human proteins in DrugBank, and between 315 514 chemicals and 9457 human proteins in the STITCH database. In addition, QuartataWeb links targets to KEGG pathways and GO annotations, completing the bridge from drugs/chemicals to function via protein targets and cellular pathways. It allows users to query a series of chemicals, drug combinations or multiple targets, to enable multi-drug, multi-target, multi-pathway analyses, toward facilitating the design of polypharmacological treatments for complex diseases. AVAILABILITY AND IMPLEMENTATION QuartataWeb is freely accessible at http://quartata.csb.pitt.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hongchun Li
- Department of Computational and Systems Biology School of Medicine.,Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fen Pei
- Department of Computational and Systems Biology School of Medicine.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Department of Computational and Systems Biology School of Medicine.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology School of Medicine.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
18
|
Kumar P, Sharma D. A potential energy and mutual information based link prediction approach for bipartite networks. Sci Rep 2020; 10:20659. [PMID: 33244025 PMCID: PMC7691373 DOI: 10.1038/s41598-020-77364-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Link prediction in networks has applications in computer science, graph theory, biology, economics, etc. Link prediction is a very well studied problem. Out of all the different versions, link prediction for unipartite graphs has attracted most attention. In this work we focus on link prediction for bipartite graphs that is based on two very important concepts-potential energy and mutual information. In the three step approach; first the bipartite graph is converted into a unipartite graph with the help of a weighted projection, next the potential energy and mutual information between each node pair in the projected graph is computed. Finally, we present Potential Energy-Mutual Information based similarity metric which helps in prediction of potential links. To evaluate the performance of the proposed algorithm four similarity metrics, namely AUC, Precision, Prediction-power and Precision@K were calculated and compared with eleven baseline algorithms. The Experimental results show that the proposed method outperforms the baseline algorithms.
Collapse
Affiliation(s)
- Purushottam Kumar
- Department of Computer Science and Engineering, Shiv Nadar University, Gautam Buddha Nagar, 201314, Uttar Pradesh, India
| | - Dolly Sharma
- Department of Computer Science and Engineering, Shiv Nadar University, Gautam Buddha Nagar, 201314, Uttar Pradesh, India.
| |
Collapse
|
19
|
He S, Wen Y, Yang X, Liu Z, Song X, Huang X, Bo X. PIMD: An Integrative Approach for Drug Repositioning Using Multiple Characterization Fusion. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:565-581. [PMID: 33075523 PMCID: PMC8377380 DOI: 10.1016/j.gpb.2018.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/21/2018] [Accepted: 10/10/2018] [Indexed: 11/28/2022]
Abstract
The accumulation of various types of drug informatics data and computational approaches for drug repositioning can accelerate pharmaceutical research and development. However, the integration of multi-dimensional drug data for precision repositioning remains a pressing challenge. Here, we propose a systematic framework named PIMD to predict drug therapeutic properties by integrating multi-dimensional data for drug repositioning. In PIMD, drug similarity networks (DSNs) based on chemical, pharmacological, and clinical data are fused into an integrated DSN (iDSN) composed of many clusters. Rather than simple fusion, PIMD offers a systematic way to annotate clusters. Unexpected drugs within clusters and drug pairs with a high iDSN similarity score are therefore identified to predict novel therapeutic uses. PIMD provides new insights into the universality, individuality, and complementarity of different drug properties by evaluating the contribution of each property data. To test the performance of PIMD, we use chemical, pharmacological, and clinical properties to generate an iDSN. Analyses of the contributions of each drug property indicate that this iDSN was driven by all data types and performs better than other DSNs. Within the top 20 recommended drug pairs, 7 drugs have been reported to be repurposed. The source code for PIMD is available at https://github.com/Sepstar/PIMD/.
Collapse
Affiliation(s)
- Song He
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yuqi Wen
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaoxi Yang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhen Liu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xinyu Song
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xin Huang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
20
|
Wen Z, Yan C, Duan G, Li S, Wu FX, Wang J. A survey on predicting microbe-disease associations: biological data and computational methods. Brief Bioinform 2020; 22:5881365. [PMID: 34020541 DOI: 10.1093/bib/bbaa157] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Various microbes have proved to be closely related to the pathogenesis of human diseases. While many computational methods for predicting human microbe-disease associations (MDAs) have been developed, few systematic reviews on these methods have been reported. In this study, we provide a comprehensive overview of the existing methods. Firstly, we introduce the data used in existing MDA prediction methods. Secondly, we classify those methods into different categories by their nature and describe their algorithms and strategies in detail. Next, experimental evaluations are conducted on representative methods using different similarity data and calculation methods to compare their prediction performances. Based on the principles of computational methods and experimental results, we discuss the advantages and disadvantages of those methods and propose suggestions for the improvement of prediction performances. Considering the problems of the MDA prediction at present stage, we discuss future work from three perspectives including data, methods and formulations at the end.
Collapse
Affiliation(s)
- Zhongqi Wen
- Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering at Central South University, Hunan, China
| | - Cheng Yan
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Guihua Duan
- School of Computer Science and Engineering, Central South University
| | - Suning Li
- Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Fang-Xiang Wu
- College of Engineering and the Department of Computer Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Jianxin Wang
- Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering at Central South University, Hunan, China
| |
Collapse
|
21
|
Dhillon BK, Smith M, Baghela A, Lee AHY, Hancock REW. Systems Biology Approaches to Understanding the Human Immune System. Front Immunol 2020; 11:1683. [PMID: 32849587 PMCID: PMC7406790 DOI: 10.3389/fimmu.2020.01683] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022] Open
Abstract
Systems biology is an approach to interrogate complex biological systems through large-scale quantification of numerous biomolecules. The immune system involves >1,500 genes/proteins in many interconnected pathways and processes, and a systems-level approach is critical in broadening our understanding of the immune response to vaccination. Changes in molecular pathways can be detected using high-throughput omics datasets (e.g., transcriptomics, proteomics, and metabolomics) by using methods such as pathway enrichment, network analysis, machine learning, etc. Importantly, integration of multiple omic datasets is becoming key to revealing novel biological insights. In this perspective article, we highlight the use of protein-protein interaction (PPI) networks as a multi-omics integration approach to unravel information flow and mechanisms during complex biological events, with a focus on the immune system. This involves a combination of tools, including: InnateDB, a database of curated interactions between genes and protein products involved in the innate immunity; NetworkAnalyst, a visualization and analysis platform for InnateDB interactions; and MetaBridge, a tool to integrate metabolite data into PPI networks. The application of these systems techniques is demonstrated for a variety of biological questions, including: the developmental trajectory of neonates during the first week of life, mechanisms in host-pathogen interaction, disease prognosis, biomarker discovery, and drug discovery and repurposing. Overall, systems biology analyses of omics data have been applied to a variety of immunology-related questions, and here we demonstrate the numerous ways in which PPI network analysis can be a powerful tool in contributing to our understanding of the immune system and the study of vaccines.
Collapse
Affiliation(s)
- Bhavjinder K. Dhillon
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Maren Smith
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Arjun Baghela
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy H. Y. Lee
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Molecular Biology & Biochemistry Department, Simon Fraser University, Burnaby, BC, Canada
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
22
|
Roy H, Nandi S. In-Silico Modeling in Drug Metabolism and Interaction: Current Strategies of Lead Discovery. Curr Pharm Des 2020; 25:3292-3305. [PMID: 31481001 DOI: 10.2174/1381612825666190903155935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Drug metabolism is a complex mechanism of human body systems to detoxify foreign particles, chemicals, and drugs through bio alterations. It involves many biochemical reactions carried out by invivo enzyme systems present in the liver, kidney, intestine, lungs, and plasma. After drug administration, it crosses several biological membranes to reach into the target site for binding and produces the therapeutic response. After that, it may undergo detoxification and excretion to get rid of the biological systems. Most of the drugs and its metabolites are excreted through kidney via urination. Some drugs and their metabolites enter into intestinal mucosa and excrete through feces. Few of the drugs enter into hepatic circulation where they go into the intestinal tract. The drug leaves the liver via the bile duct and is excreted through feces. Therefore, the study of total methodology of drug biotransformation and interactions with various targets is costly. METHODS To minimize time and cost, in-silico algorithms have been utilized for lead-like drug discovery. Insilico modeling is the process where a computer model with a suitable algorithm is developed to perform a controlled experiment. It involves the combination of both in-vivo and in-vitro experimentation with virtual trials, eliminating the non-significant variables from a large number of variable parameters. Whereas, the major challenge for the experimenter is the selection and validation of the preferred model, as well as precise simulation in real physiological status. RESULTS The present review discussed the application of in-silico models to predict absorption, distribution, metabolism, and excretion (ADME) properties of drug molecules and also access the net rate of metabolism of a compound. CONCLUSION It helps with the identification of enzyme isoforms; which are likely to metabolize a compound, as well as the concentration dependence of metabolism and the identification of expected metabolites. In terms of drug-drug interactions (DDIs), models have been described for the inhibition of metabolism of one compound by another, and for the compound-dependent induction of drug-metabolizing enzymes.
Collapse
Affiliation(s)
- Harekrishna Roy
- Nirmala College of Pharmacy, Mangalagiri, Guntur, Affiliated to Acharya Nagarjuna University, Andhra Pradesh-522503, India
| | - Sisir Nandi
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research, Affiliated to Uttarakhand Technical University, Kashipur-244713, India
| |
Collapse
|
23
|
Kaushik AC, Mehmood A, Dai X, Wei DQ. A comparative chemogenic analysis for predicting Drug-Target Pair via Machine Learning Approaches. Sci Rep 2020; 10:6870. [PMID: 32322011 PMCID: PMC7176722 DOI: 10.1038/s41598-020-63842-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/04/2020] [Indexed: 12/26/2022] Open
Abstract
A computational technique for predicting the DTIs has now turned out to be an indispensable job during the process of drug finding. It tapers the exploration room for interactions by propounding possible interaction contenders for authentication through experiments of wet-lab which are known for their expensiveness and time consumption. Chemogenomics, an emerging research area focused on the systematic examination of the biological impact of a broad series of minute molecular-weighting ligands on a broad raiment of macromolecular target spots. Additionally, with the advancement in time, the complexity of the algorithms is increasing which may result in the entry of big data technologies like Spark in this field soon. In the presented work, we intend to offer an inclusive idea and realistic evaluation of the computational Drug Target Interaction projection approaches, to perform as a guide and reference for researchers who are carrying out work in a similar direction. Precisely, we first explain the data utilized in computational Drug Target Interaction prediction attempts like this. We then sort and explain the best and most modern techniques for the prediction of DTIs. Then, a realistic assessment is executed to show the projection performance of several illustrative approaches in various situations. Ultimately, we underline possible opportunities for additional improvement of Drug Target Interaction projection enactment and also linked study objectives.
Collapse
Affiliation(s)
- Aman Chandra Kaushik
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Aamir Mehmood
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
24
|
Bagherian M, Sabeti E, Wang K, Sartor MA, Nikolovska-Coleska Z, Najarian K. Machine learning approaches and databases for prediction of drug-target interaction: a survey paper. Brief Bioinform 2020; 22:247-269. [PMID: 31950972 PMCID: PMC7820849 DOI: 10.1093/bib/bbz157] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
The task of predicting the interactions between drugs and targets plays a key role in the process of drug discovery. There is a need to develop novel and efficient prediction approaches in order to avoid costly and laborious yet not-always-deterministic experiments to determine drug–target interactions (DTIs) by experiments alone. These approaches should be capable of identifying the potential DTIs in a timely manner. In this article, we describe the data required for the task of DTI prediction followed by a comprehensive catalog consisting of machine learning methods and databases, which have been proposed and utilized to predict DTIs. The advantages and disadvantages of each set of methods are also briefly discussed. Lastly, the challenges one may face in prediction of DTI using machine learning approaches are highlighted and we conclude by shedding some lights on important future research directions.
Collapse
Affiliation(s)
- Maryam Bagherian
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Elyas Sabeti
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kai Wang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Maureen A Sartor
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Kayvan Najarian
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
25
|
Sarsaiya S, Shi J, Chen J. Bioengineering tools for the production of pharmaceuticals: current perspective and future outlook. Bioengineered 2019; 10:469-492. [PMID: 31656120 PMCID: PMC6844412 DOI: 10.1080/21655979.2019.1682108] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/08/2019] [Accepted: 10/11/2019] [Indexed: 01/18/2023] Open
Abstract
The bioengineering tools have significant advantages through less time-consuming and utilized as a promising stage for the production of pharmaceutical bioproducts under the single platform. This review highlighted the advantages and current improvement in the plant, animal and microbial bioengineering tools and outlines feasible approaches by biological and process's bioengineering levels for advancing the economic feasibility of pharmaceutical's production. The critical analysis results revealed that system biology and synthetic biology along with advanced bioengineering tools like transcriptome, proteome, metabolome and nano bioengineering tools have shown a promising impact on the development of pharmaceutical's bioproducts. Tools to overcome and resolve the accompanying encounters of pharmaceutical's production that include nano bioengineering tools are also discussed. As a summary and prospect, it also gives new insight into the challenges and possible breakthrough of the development of pharmaceutical's bioproducts through bioengineering tools.
Collapse
Affiliation(s)
- Surendra Sarsaiya
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Bioresource Institute for Healthy Utilization, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jishuang Chen
- Bioresource Institute for Healthy Utilization, Zunyi Medical University, Zunyi, China
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| |
Collapse
|
26
|
Drug Side-Effect Prediction Via Random Walk on the Signed Heterogeneous Drug Network. MOLECULES (BASEL, SWITZERLAND) 2019; 24:molecules24203668. [PMID: 31614686 PMCID: PMC6832386 DOI: 10.3390/molecules24203668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 11/30/2022]
Abstract
Drug side-effects have become a major public health concern as they are the underlying cause of over a million serious injuries and deaths each year. Therefore, it is of critical importance to detect side-effects as early as possible. Existing computational methods mainly utilize the drug chemical profile and the drug biological profile to predict the side-effects of a drug. In the utilized drug biological profile information, they only focus on drug–target interactions and neglect the modes of action of drugs on target proteins. In this paper, we develop a new method for predicting potential side-effects of drugs based on more comprehensive drug information in which the modes of action of drugs on target proteins are integrated. Drug information of multiple types is modeled as a signed heterogeneous information network. We propose a signed heterogeneous information network embedding framework for learning drug embeddings and predicting side-effects of drugs. We use two bias random walk procedures to obtain drug sequences and train a Skip-gram model to learn drug embeddings. We experimentally demonstrate the performance of the proposed method by comparison with state-of-the-art methods. Furthermore, the results of a case study support our hypothesis that modes of action of drugs on target proteins are meaningful in side-effect prediction.
Collapse
|
27
|
Durán C, Daminelli S, Thomas JM, Haupt VJ, Schroeder M, Cannistraci CV. Pioneering topological methods for network-based drug-target prediction by exploiting a brain-network self-organization theory. Brief Bioinform 2019; 19:1183-1202. [PMID: 28453640 PMCID: PMC6291778 DOI: 10.1093/bib/bbx041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Indexed: 01/03/2023] Open
Abstract
The bipartite network representation of the drug–target interactions (DTIs) in a biosystem enhances understanding of the drugs’ multifaceted action modes, suggests therapeutic switching for approved drugs and unveils possible side effects. As experimental testing of DTIs is costly and time-consuming, computational predictors are of great aid. Here, for the first time, state-of-the-art DTI supervised predictors custom-made in network biology were compared—using standard and innovative validation frameworks—with unsupervised pure topological-based models designed for general-purpose link prediction in bipartite networks. Surprisingly, our results show that the bipartite topology alone, if adequately exploited by means of the recently proposed local-community-paradigm (LCP) theory—initially detected in brain-network topological self-organization and afterwards generalized to any complex network—is able to suggest highly reliable predictions, with comparable performance with the state-of-the-art-supervised methods that exploit additional (non-topological, for instance biochemical) DTI knowledge. Furthermore, a detailed analysis of the novel predictions revealed that each class of methods prioritizes distinct true interactions; hence, combining methodologies based on diverse principles represents a promising strategy to improve drug–target discovery. To conclude, this study promotes the power of bio-inspired computing, demonstrating that simple unsupervised rules inspired by principles of topological self-organization and adaptiveness arising during learning in living intelligent systems (like the brain) can efficiently equal perform complicated algorithms based on advanced, supervised and knowledge-based engineering.
Collapse
Affiliation(s)
| | - Simone Daminelli
- Corresponding authors: Carlo Cannistraci, Biomedical Cybernetics Group at Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Department of Physics, Technische Universität Dresden (TUD), Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40080; E-mail: ; Simone Daminelli, Bioinformatics Group at Biotechnology Center (BIOTEC), Center of Molecular Cellular Bioengineering (CMCB), TUD, Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40060; E-mail: ; Michael Schroeder, Bioinformatics Group at Biotechnology Center (BIOTEC), Center of Molecular and Cellular Bioengineering (CMCB), TUD Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40062; E-mail:
| | | | | | - Michael Schroeder
- Corresponding authors: Carlo Cannistraci, Biomedical Cybernetics Group at Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Department of Physics, Technische Universität Dresden (TUD), Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40080; E-mail: ; Simone Daminelli, Bioinformatics Group at Biotechnology Center (BIOTEC), Center of Molecular Cellular Bioengineering (CMCB), TUD, Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40060; E-mail: ; Michael Schroeder, Bioinformatics Group at Biotechnology Center (BIOTEC), Center of Molecular and Cellular Bioengineering (CMCB), TUD Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40062; E-mail:
| | - Carlo Vittorio Cannistraci
- Corresponding authors: Carlo Cannistraci, Biomedical Cybernetics Group at Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Department of Physics, Technische Universität Dresden (TUD), Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40080; E-mail: ; Simone Daminelli, Bioinformatics Group at Biotechnology Center (BIOTEC), Center of Molecular Cellular Bioengineering (CMCB), TUD, Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40060; E-mail: ; Michael Schroeder, Bioinformatics Group at Biotechnology Center (BIOTEC), Center of Molecular and Cellular Bioengineering (CMCB), TUD Tatzberg 47-49, 01307 Dresden, Germany, Tel.: +49 (0)351 463 40062; E-mail:
| |
Collapse
|
28
|
Zhou L, Li Z, Yang J, Tian G, Liu F, Wen H, Peng L, Chen M, Xiang J, Peng L. Revealing Drug-Target Interactions with Computational Models and Algorithms. Molecules 2019; 24:molecules24091714. [PMID: 31052598 PMCID: PMC6540161 DOI: 10.3390/molecules24091714] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/02/2022] Open
Abstract
Background: Identifying possible drug-target interactions (DTIs) has become an important task in drug research and development. Although high-throughput screening is becoming available, experimental methods narrow down the validation space because of extremely high cost, low success rate, and time consumption. Therefore, various computational models have been exploited to infer DTI candidates. Methods: We introduced relevant databases and packages, mainly provided a comprehensive review of computational models for DTI identification, including network-based algorithms and machine learning-based methods. Specially, machine learning-based methods mainly include bipartite local model, matrix factorization, regularized least squares, and deep learning. Results: Although computational methods have obtained significant improvement in the process of DTI prediction, these models have their limitations. We discussed potential avenues for boosting DTI prediction accuracy as well as further directions.
Collapse
Affiliation(s)
- Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| | - Zejun Li
- School of Computer Science, Hunan Institute of Technology, Henyang 421002, Hunan, China.
| | | | - Geng Tian
- Geneis (Beijing) Co. Ltd., Beijing 100102, China.
| | - Fuxing Liu
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| | - Hong Wen
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| | - Li Peng
- School of Computer Science, University of Science and Technology of Hunan, Xiangtan 411201, Hunan, China.
| | - Min Chen
- School of Computer Science, Hunan Institute of Technology, Henyang 421002, Hunan, China.
| | - Ju Xiang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China.
- Neuroscience Research Center, Department of Basic Medical Sciences, Changsha Medical University, Changsha 410219, Hunan, China.
| | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| |
Collapse
|
29
|
Predicting drug-target interaction network using deep learning model. Comput Biol Chem 2019; 80:90-101. [PMID: 30939415 DOI: 10.1016/j.compbiolchem.2019.03.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/23/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Traditional methods for drug discovery are time-consuming and expensive, so efforts are being made to repurpose existing drugs. To find new ways for drug repurposing, many computational approaches have been proposed to predict drug-target interactions (DTIs). However, due to the high-dimensional nature of the data sets extracted from drugs and targets, traditional machine learning approaches, such as logistic regression analysis, cannot analyze these data sets efficiently. To overcome this issue, we propose LASSO (Least absolute shrinkage and selection operator)-based regularized linear classification models and a LASSO-DNN (Deep Neural Network) model based on LASSO feature selection to predict DTIs. These methods are demonstrated for repurposing drugs for breast cancer treatment. METHODS We collected drug descriptors, protein sequence data from Drugbank and protein domain information from NCBI. Validated DTIs were downloaded from Drugbank. A new similarity-based approach was developed to build the negative DTIs. We proposed multiple LASSO models to integrate different combinations of feature sets to explore the prediction power and predict DTIs. Furthermore, building on the features extracted from the LASSO models with the best performance, we also introduced a LASSO-DNN model to predict DTIs. The performance of our newly proposed DNN model (LASSO-DNN) was compared with the LASSO, standard logistic (SLG) regression, support vector machine (SVM), and standard DNN models. RESULTS Experimental results showed that the LASSO-DNN over performed the SLG, LASSO, SVM and standard DNN models. In particular, the LASSO models with protein tripeptide composition (TC) features and domain features were superior to those that contained other protein information, which may imply that TC and domain information could be better representations of proteins. Furthermore, we showed that the top ranked DTIs predicted using the LASSO-DNN model can potentially be used for repurposing existing drugs for breast cancer based on risk gene information. CONCLUSIONS In summary, we demonstrated that the efficient representations of drug and target features are key for building learning models for predicting DTIs. The disease-associated risk genes identified from large-scale genomic studies are the potential drug targets, which can be used for drug repurposing.
Collapse
|
30
|
Sydow D, Burggraaff L, Szengel A, van Vlijmen HWT, IJzerman AP, van Westen GJP, Volkamer A. Advances and Challenges in Computational Target Prediction. J Chem Inf Model 2019; 59:1728-1742. [DOI: 10.1021/acs.jcim.8b00832] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Dominique Sydow
- In silico Toxicology, Institute of Physiology, Charité − Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Lindsey Burggraaff
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Angelika Szengel
- In silico Toxicology, Institute of Physiology, Charité − Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Herman W. T. van Vlijmen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
- Computational Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Adriaan P. IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Gerard J. P. van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Andrea Volkamer
- In silico Toxicology, Institute of Physiology, Charité − Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
31
|
Zheng Y, Peng H, Ghosh S, Lan C, Li J. Inverse similarity and reliable negative samples for drug side-effect prediction. BMC Bioinformatics 2019; 19:554. [PMID: 30717666 PMCID: PMC7402513 DOI: 10.1186/s12859-018-2563-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 12/07/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND In silico prediction of potential drug side-effects is of crucial importance for drug development, since wet experimental identification of drug side-effects is expensive and time-consuming. Existing computational methods mainly focus on leveraging validated drug side-effect relations for the prediction. The performance is severely impeded by the lack of reliable negative training data. Thus, a method to select reliable negative samples becomes vital in the performance improvement. METHODS Most of the existing computational prediction methods are essentially based on the assumption that similar drugs are inclined to share the same side-effects, which has given rise to remarkable performance. It is also rational to assume an inverse proposition that dissimilar drugs are less likely to share the same side-effects. Based on this inverse similarity hypothesis, we proposed a novel method to select highly-reliable negative samples for side-effect prediction. The first step of our method is to build a drug similarity integration framework to measure the similarity between drugs from different perspectives. This step integrates drug chemical structures, drug target proteins, drug substituents, and drug therapeutic information as features into a unified framework. Then, a similarity score between each candidate negative drug and validated positive drugs is calculated using the similarity integration framework. Those candidate negative drugs with lower similarity scores are preferentially selected as negative samples. Finally, both the validated positive drugs and the selected highly-reliable negative samples are used for predictions. RESULTS The performance of the proposed method was evaluated on simulative side-effect prediction of 917 DrugBank drugs, comparing with four machine-learning algorithms. Extensive experiments show that the drug similarity integration framework has superior capability in capturing drug features, achieving much better performance than those based on a single type of drug property. Besides, the four machine-learning algorithms achieved significant improvement in macro-averaging F1-score (e.g., SVM from 0.655 to 0.898), macro-averaging precision (e.g., RBF from 0.592 to 0.828) and macro-averaging recall (e.g., KNN from 0.651 to 0.772) complimentarily attributed to the highly-reliable negative samples selected by the proposed method. CONCLUSIONS The results suggest that the inverse similarity hypothesis and the integration of different drug properties are valuable for side-effect prediction. The selection of highly-reliable negative samples can also make significant contributions to the performance improvement.
Collapse
Affiliation(s)
- Yi Zheng
- Advanced Analytics Institute, FEIT, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Hui Peng
- Advanced Analytics Institute, FEIT, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Shameek Ghosh
- Advanced Analytics Institute, FEIT, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Chaowang Lan
- Advanced Analytics Institute, FEIT, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Jinyan Li
- Advanced Analytics Institute, FEIT, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia.
| |
Collapse
|
32
|
Taylor DL, Gough A, Schurdak ME, Vernetti L, Chennubhotla CS, Lefever D, Pei F, Faeder JR, Lezon TR, Stern AM, Bahar I. Harnessing Human Microphysiology Systems as Key Experimental Models for Quantitative Systems Pharmacology. Handb Exp Pharmacol 2019; 260:327-367. [PMID: 31201557 PMCID: PMC6911651 DOI: 10.1007/164_2019_239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Two technologies that have emerged in the last decade offer a new paradigm for modern pharmacology, as well as drug discovery and development. Quantitative systems pharmacology (QSP) is a complementary approach to traditional, target-centric pharmacology and drug discovery and is based on an iterative application of computational and systems biology methods with multiscale experimental methods, both of which include models of ADME-Tox and disease. QSP has emerged as a new approach due to the low efficiency of success in developing therapeutics based on the existing target-centric paradigm. Likewise, human microphysiology systems (MPS) are experimental models complementary to existing animal models and are based on the use of human primary cells, adult stem cells, and/or induced pluripotent stem cells (iPSCs) to mimic human tissues and organ functions/structures involved in disease and ADME-Tox. Human MPS experimental models have been developed to address the relatively low concordance of human disease and ADME-Tox with engineered, experimental animal models of disease. The integration of the QSP paradigm with the use of human MPS has the potential to enhance the process of drug discovery and development.
Collapse
Affiliation(s)
- D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark E Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chakra S Chennubhotla
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Lefever
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Fen Pei
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Faeder
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy R Lezon
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Abstract
Pharmacological science is trying to establish the link between chemicals, targets, and disease-related phenotypes. A plethora of chemical proteomics and structural data have been generated, thanks to the target-based approach that has dominated drug discovery at the turn of the century. There is an invaluable source of information for in silico target profiling. Prediction is based on the principle of chemical similarity (similar drugs bind similar targets) or on first principles from the biophysics of molecular interactions. In the first case, compound comparison is made through ligand-based chemical similarity search or through classifier-based machine learning approach. The 3D techniques are based on 3D structural descriptors or energy-based scoring scheme to infer a binding affinity of a compound with its putative target. More recently, a new approach based on compound set metric has been proposed in which a query compound is compared with a whole of compounds associated with a target or a family of targets. This chapter reviews the different techniques of in silico target profiling and their main applications such as inference of unwanted targets, drug repurposing, or compound prioritization after phenotypic-based screening campaigns.
Collapse
|
34
|
Abstract
Drugs modulate disease states through their actions on targets in the body. Determining these targets aids the focused development of new treatments, and helps to better characterize those already employed. One means of accomplishing this is through the deployment of in silico methodologies, harnessing computational analytical and predictive power to produce educated hypotheses for experimental verification. Here, we provide an overview of the current state of the art, describe some of the well-established methods in detail, and reflect on how they, and emerging technologies promoting the incorporation of complex and heterogeneous data-sets, can be employed to improve our understanding of (poly)pharmacology.
Collapse
Affiliation(s)
- Ryan Byrne
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.
| |
Collapse
|
35
|
Su C, Tong J, Zhu Y, Cui P, Wang F. Network embedding in biomedical data science. Brief Bioinform 2018; 21:182-197. [PMID: 30535359 DOI: 10.1093/bib/bby117] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/08/2018] [Accepted: 11/03/2018] [Indexed: 12/15/2022] Open
Abstract
AbstractOwning to the rapid development of computer technologies, an increasing number of relational data have been emerging in modern biomedical research. Many network-based learning methods have been proposed to perform analysis on such data, which provide people a deep understanding of topology and knowledge behind the biomedical networks and benefit a lot of applications for human healthcare. However, most network-based methods suffer from high computational and space cost. There remain challenges on handling high dimensionality and sparsity of the biomedical networks. The latest advances in network embedding technologies provide new effective paradigms to solve the network analysis problem. It converts network into a low-dimensional space while maximally preserves structural properties. In this way, downstream tasks such as link prediction and node classification can be done by traditional machine learning methods. In this survey, we conduct a comprehensive review of the literature on applying network embedding to advance the biomedical domain. We first briefly introduce the widely used network embedding models. After that, we carefully discuss how the network embedding approaches were performed on biomedical networks as well as how they accelerated the downstream tasks in biomedical science. Finally, we discuss challenges the existing network embedding applications in biomedical domains are faced with and suggest several promising future directions for a better improvement in human healthcare.
Collapse
Affiliation(s)
- Chang Su
- Department of Healthcare Policy and Research, Weill Cornell Medicine at Cornell University, New York, NY, USA
| | - Jie Tong
- Department of Mechanical and Aerospace Engineering at New York University, New York, NY, USA
| | - Yongjun Zhu
- Department of Library and Information Science, Sungkyunkwan University, Seoul, South Korea
| | - Peng Cui
- Department of Computer Science and Technology, Tsinghua University, Beijing, China
| | - Fei Wang
- Department of Healthcare Policy and Research, Weill Cornell Medicine at Cornell University, New York, NY, USA
| |
Collapse
|
36
|
Van Vleet TR, Liguori MJ, Lynch JJ, Rao M, Warder S. Screening Strategies and Methods for Better Off-Target Liability Prediction and Identification of Small-Molecule Pharmaceuticals. SLAS DISCOVERY 2018; 24:1-24. [PMID: 30196745 DOI: 10.1177/2472555218799713] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pharmaceutical discovery and development is a long and expensive process that, unfortunately, still results in a low success rate, with drug safety continuing to be a major impedance. Improved safety screening strategies and methods are needed to more effectively fill this critical gap. Recent advances in informatics are now making it possible to manage bigger data sets and integrate multiple sources of screening data in a manner that can potentially improve the selection of higher-quality drug candidates. Integrated screening paradigms have become the norm in Pharma, both in discovery screening and in the identification of off-target toxicity mechanisms during later-stage development. Furthermore, advances in computational methods are making in silico screens more relevant and suggest that they may represent a feasible option for augmenting the current screening paradigm. This paper outlines several fundamental methods of the current drug screening processes across Pharma and emerging techniques/technologies that promise to improve molecule selection. In addition, the authors discuss integrated screening strategies and provide examples of advanced screening paradigms.
Collapse
Affiliation(s)
- Terry R Van Vleet
- 1 Department of Investigative Toxicology and Pathology, AbbVie, N Chicago, IL, USA
| | - Michael J Liguori
- 1 Department of Investigative Toxicology and Pathology, AbbVie, N Chicago, IL, USA
| | - James J Lynch
- 2 Department of Integrated Science and Technology, AbbVie, N Chicago, IL, USA
| | - Mohan Rao
- 1 Department of Investigative Toxicology and Pathology, AbbVie, N Chicago, IL, USA
| | - Scott Warder
- 3 Department of Target Enabling Science and Technology, AbbVie, N Chicago, IL, USA
| |
Collapse
|
37
|
Wang C, Kurgan L. Review and comparative assessment of similarity-based methods for prediction of drug–protein interactions in the druggable human proteome. Brief Bioinform 2018; 20:2066-2087. [DOI: 10.1093/bib/bby069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 12/18/2022] Open
Abstract
AbstractDrug–protein interactions (DPIs) underlie the desired therapeutic actions and the adverse side effects of a significant majority of drugs. Computational prediction of DPIs facilitates research in drug discovery, characterization and repurposing. Similarity-based methods that do not require knowledge of protein structures are particularly suitable for druggable genome-wide predictions of DPIs. We review 35 high-impact similarity-based predictors that were published in the past decade. We group them based on three types of similarities and their combinations that they use. We discuss and compare key aspects of these methods including source databases, internal databases and their predictive models. Using our novel benchmark database, we perform comparative empirical analysis of predictive performance of seven types of representative predictors that utilize each type of similarity individually and all possible combinations of similarities. We assess predictive quality at the database-wide DPI level and we are the first to also include evaluation over individual drugs. Our comprehensive analysis shows that predictors that use more similarity types outperform methods that employ fewer similarities, and that the model combining all three types of similarities secures area under the receiver operating characteristic curve of 0.93. We offer a comprehensive analysis of sensitivity of predictive performance to intrinsic and extrinsic characteristics of the considered predictors. We find that predictive performance is sensitive to low levels of similarities between sequences of the drug targets and several extrinsic properties of the input drug structures, drug profiles and drug targets. The benchmark database and a webserver for the seven predictors are freely available at http://biomine.cs.vcu.edu/servers/CONNECTOR/.
Collapse
Affiliation(s)
- Chen Wang
- Computer Science Department, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Lukasz Kurgan
- Computer Science Department, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
38
|
Huang G, Li J, Zhao C. Computational Prediction and Analysis of Associations between Small Molecules and Binding-Associated S-Nitrosylation Sites. Molecules 2018; 23:molecules23040954. [PMID: 29671802 PMCID: PMC6017196 DOI: 10.3390/molecules23040954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/30/2018] [Accepted: 04/09/2018] [Indexed: 01/12/2023] Open
Abstract
Interactions between drugs and proteins occupy a central position during the process of drug discovery and development. Numerous methods have recently been developed for identifying drug–target interactions, but few have been devoted to finding interactions between post-translationally modified proteins and drugs. We presented a machine learning-based method for identifying associations between small molecules and binding-associated S-nitrosylated (SNO-) proteins. Namely, small molecules were encoded by molecular fingerprint, SNO-proteins were encoded by the information entropy-based method, and the random forest was used to train a classifier. Ten-fold and leave-one-out cross validations achieved, respectively, 0.7235 and 0.7490 of the area under a receiver operating characteristic curve. Computational analysis of similarity suggested that SNO-proteins associated with the same drug shared statistically significant similarity, and vice versa. This method and finding are useful to identify drug–SNO associations and further facilitate the discovery and development of SNO-associated drugs.
Collapse
Affiliation(s)
- Guohua Huang
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang 422000, China.
- College of Information Engineering, Shaoyang University, Shaoyang 422000, China.
| | - Jincheng Li
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang 422000, China.
- College of Information Engineering, Shaoyang University, Shaoyang 422000, China.
| | - Chenglin Zhao
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang 422000, China.
- College of Information Engineering, Shaoyang University, Shaoyang 422000, China.
| |
Collapse
|
39
|
Ezzat A, Wu M, Li XL, Kwoh CK. Computational prediction of drug–target interactions using chemogenomic approaches: an empirical survey. Brief Bioinform 2018; 20:1337-1357. [DOI: 10.1093/bib/bby002] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/21/2017] [Indexed: 01/18/2023] Open
Abstract
Abstract
Computational prediction of drug–target interactions (DTIs) has become an essential task in the drug discovery process. It narrows down the search space for interactions by suggesting potential interaction candidates for validation via wet-lab experiments that are well known to be expensive and time-consuming. In this article, we aim to provide a comprehensive overview and empirical evaluation on the computational DTI prediction techniques, to act as a guide and reference for our fellow researchers. Specifically, we first describe the data used in such computational DTI prediction efforts. We then categorize and elaborate the state-of-the-art methods for predicting DTIs. Next, an empirical comparison is performed to demonstrate the prediction performance of some representative methods under different scenarios. We also present interesting findings from our evaluation study, discussing the advantages and disadvantages of each method. Finally, we highlight potential avenues for further enhancement of DTI prediction performance as well as related research directions.
Collapse
|
40
|
Zhu Y, Elemento O, Pathak J, Wang F. Drug knowledge bases and their applications in biomedical informatics research. Brief Bioinform 2018; 20:1308-1321. [DOI: 10.1093/bib/bbx169] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/15/2017] [Indexed: 11/14/2022] Open
Abstract
Abstract
Recent advances in biomedical research have generated a large volume of drug-related data. To effectively handle this flood of data, many initiatives have been taken to help researchers make good use of them. As the results of these initiatives, many drug knowledge bases have been constructed. They range from simple ones with specific focuses to comprehensive ones that contain information on almost every aspect of a drug. These curated drug knowledge bases have made significant contributions to the development of efficient and effective health information technologies for better health-care service delivery. Understanding and comparing existing drug knowledge bases and how they are applied in various biomedical studies will help us recognize the state of the art and design better knowledge bases in the future. In addition, researchers can get insights on novel applications of the drug knowledge bases through a review of successful use cases. In this study, we provide a review of existing popular drug knowledge bases and their applications in drug-related studies. We discuss challenges in constructing and using drug knowledge bases as well as future research directions toward a better ecosystem of drug knowledge bases.
Collapse
|
41
|
Wang W, Chen X, Jiao P, Jin D. Similarity-based Regularized Latent Feature Model for Link Prediction in Bipartite Networks. Sci Rep 2017; 7:16996. [PMID: 29208988 PMCID: PMC5717264 DOI: 10.1038/s41598-017-17157-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/21/2017] [Indexed: 01/05/2023] Open
Abstract
Link prediction is an attractive research topic in the field of data mining and has significant applications in improving performance of recommendation system and exploring evolving mechanisms of the complex networks. A variety of complex systems in real world should be abstractly represented as bipartite networks, in which there are two types of nodes and no links connect nodes of the same type. In this paper, we propose a framework for link prediction in bipartite networks by combining the similarity based structure and the latent feature model from a new perspective. The framework is called Similarity Regularized Nonnegative Matrix Factorization (SRNMF), which explicitly takes the local characteristics into consideration and encodes the geometrical information of the networks by constructing a similarity based matrix. We also develop an iterative scheme to solve the objective function based on gradient descent. Extensive experiments on a variety of real world bipartite networks show that the proposed framework of link prediction has a more competitive, preferable and stable performance in comparison with the state-of-art methods.
Collapse
Affiliation(s)
- Wenjun Wang
- School of Computer Science and Technology, Tianjin University, Tianjin, 300354, China.,Tianjin Engineering Center of SmartSafety and Bigdata Technology, Tianjin University, Tianjin, 300354, China.,Tianjin Key Laboratory of Advanced Networking (TANK), Tianjin Key Laboratory, Tianjin, 300354, China
| | - Xue Chen
- School of Computer Science and Technology, Tianjin University, Tianjin, 300354, China
| | - Pengfei Jiao
- School of Computer Science and Technology, Tianjin University, Tianjin, 300354, China.
| | - Di Jin
- School of Computer Science and Technology, Tianjin University, Tianjin, 300354, China
| |
Collapse
|
42
|
Identification of candidate drugs using tensor-decomposition-based unsupervised feature extraction in integrated analysis of gene expression between diseases and DrugMatrix datasets. Sci Rep 2017; 7:13733. [PMID: 29062063 PMCID: PMC5653784 DOI: 10.1038/s41598-017-13003-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023] Open
Abstract
Identifying drug target genes in gene expression profiles is not straightforward. Because a drug targets proteins and not mRNAs, the mRNA expression of drug target genes is not always altered. In addition, the interaction between a drug and protein can be context dependent; this means that simple drug incubation experiments on cell lines do not always reflect the real situation during active disease. In this paper, I applied tensor-decomposition-based unsupervised feature extraction to the integrated analysis using a mathematical product of gene expression in various diseases and gene expression in the DrugMatrix dataset, where comprehensive data on gene expression during various drug treatments of rats are reported. I found that this strategy, in a fully unsupervised manner, enables researchers to identify a combined set of genes and compounds that significantly overlap with gene and drug interactions identified in the past. As an example illustrating the usefulness of this strategy in drug discovery experiments, I considered cirrhosis, for which no effective drugs have ever been proposed. The present strategy identified two promising therapeutic-target genes, CYPOR and HNFA4; for their protein products, bezafibrate was identified as a promising candidate drug, supported by in silico docking analysis.
Collapse
|
43
|
Cheng T, Hao M, Takeda T, Bryant SH, Wang Y. Large-Scale Prediction of Drug-Target Interaction: a Data-Centric Review. AAPS J 2017; 19:1264-1275. [PMID: 28577120 PMCID: PMC11097213 DOI: 10.1208/s12248-017-0092-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/25/2017] [Indexed: 11/30/2022] Open
Abstract
The prediction of drug-target interactions (DTIs) is of extraordinary significance to modern drug discovery in terms of suggesting new drug candidates and repositioning old drugs. Despite technological advances, large-scale experimental determination of DTIs is still expensive and laborious. Effective and low-cost computational alternatives remain in strong need. Meanwhile, open-access resources have been rapidly growing with massive amount of bioactivity data becoming available, creating unprecedented opportunities for the development of novel in silico models for large-scale DTI prediction. In this work, we review the state-of-the-art computational approaches for identifying DTIs from a data-centric perspective: what the underlying data are and how they are utilized in each study. We also summarize popular public data resources and online tools for DTI prediction. It is found that various types of data were employed including properties of chemical structures, drug therapeutic effects and side effects, drug-target binding, drug-drug interactions, bioactivity data of drug molecules across multiple biological targets, and drug-induced gene expressions. More often, the heterogeneous data were integrated to offer better performance. However, challenges remain such as handling data imbalance, incorporating negative samples and quantitative bioactivity data, as well as maintaining cross-links among different data sources, which are essential for large-scale and automated information integration.
Collapse
Affiliation(s)
- Tiejun Cheng
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Ming Hao
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Takako Takeda
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Stephen H Bryant
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Yanli Wang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA.
| |
Collapse
|
44
|
Menezes JCJMDS, Edraki N, Kamat SP, Khoshneviszadeh M, Kayani Z, Mirzaei HH, Miri R, Erfani N, Nejati M, Cavaleiro JAS, Silva T, Saso L, Borges F, Firuzi O. Long Chain Alkyl Esters of Hydroxycinnamic Acids as Promising Anticancer Agents: Selective Induction of Apoptosis in Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7228-7239. [PMID: 28718636 DOI: 10.1021/acs.jafc.7b01388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cancer is the major cause of morbidity and mortality worldwide. Hydroxycinnamic acids (HCAs) are naturally occurring compounds and their alkyl esters may possess enhanced biological activities. We evaluated C4, C14, C16, and C18 alkyl esters of p-coumaric, ferulic, sinapic, and caffeic acids (19 compounds) for their cytotoxic activity against four human cancer cells and also examined their effect on cell cycle alteration and apoptosis induction. The tetradecyl (1c) and hexadecyl (1d) esters of p-coumaric acid and tetradecyl ester of caffeic acid (4c), but not the parental HCAs, were selectively effective against MOLT-4 (human lymphoblastic leukemia) cells with IC50 values of 0.123 ± 0.012, 0.301 ± 0.069 and 1.0 ± 0.1 μM, respectively. Compounds 1c, 1d, and 4c significantly increased apoptotic cells in sub-G1 phase and activated the caspase-3 enzyme in MOLT-4 cells. Compound 1c was 15.4 and 23.6 times more potent than doxorubicin and cisplatin, respectively, against the drug resistant MES-SA-DX5 uterine sarcoma cells. These p-coumarate esters were several times less effective against NIH/3T3 fibroblast cells. Docking studies showed that 1c may cause cytotoxicity by interaction with carbonic anhydrase IX. In conclusion, long chain alkyl esters of p-coumaric acid are promising scaffolds for selective apoptosis induction in cancer cells.
Collapse
Affiliation(s)
- José C J M D S Menezes
- Department of Chemistry & QOPNA, University of Aveiro , 3810-193 Aveiro, Portugal
- Department of Chemistry, Goa University , Taleigao 403 206 Goa India
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | | | - Mahsima Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - Zahra Kayani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - Hossein Hadavand Mirzaei
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
- Department of Molecular Physiology, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO) , Karaj, Iran
| | - Ramin Miri
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - Nasrollah Erfani
- Institute for Cancer Research (ICR), School of Medicine, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Maryam Nejati
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| | - José A S Cavaleiro
- Department of Chemistry & QOPNA, University of Aveiro , 3810-193 Aveiro, Portugal
| | - Tiago Silva
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , 4169-007 Porto, Portugal
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer″, Sapienza University of Rome , Italy
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , 4169-007 Porto, Portugal
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz, 71345-1149 Iran
| |
Collapse
|
45
|
Vilar S, Hripcsak G. The role of drug profiles as similarity metrics: applications to repurposing, adverse effects detection and drug-drug interactions. Brief Bioinform 2017; 18:670-681. [PMID: 27273288 PMCID: PMC6078166 DOI: 10.1093/bib/bbw048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/18/2016] [Indexed: 12/30/2022] Open
Abstract
Explosion of the availability of big data sources along with the development in computational methods provides a useful framework to study drugs' actions, such as interactions with pharmacological targets and off-targets. Databases related to protein interactions, adverse effects and genomic profiles are available to be used for the construction of computational models. In this article, we focus on the description of biological profiles for drugs that can be used as a system to compare similarity and create methods to predict and analyze drugs' actions. We highlight profiles constructed with different biological data, such as target-protein interactions, gene expression measurements, adverse effects and disease profiles. We focus on the discovery of new targets or pathways for drugs already in the pharmaceutical market, also called drug repurposing, in the interaction with off-targets responsible for adverse reactions and in drug-drug interaction analysis. The current and future applications, strengths and challenges facing all these methods are also discussed. Biological profiles or signatures are an important source of data generation to deeply analyze biological actions with important implications in drug-related studies.
Collapse
Affiliation(s)
- Santiago Vilar
- Corresponding author: Santiago Vilar, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail: ; George Hripcsak, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail:
| | - George Hripcsak
- Corresponding author: Santiago Vilar, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail: ; George Hripcsak, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail:
| |
Collapse
|
46
|
Humbeck L, Koch O. What Can We Learn from Bioactivity Data? Chemoinformatics Tools and Applications in Chemical Biology Research. ACS Chem Biol 2017; 12:23-35. [PMID: 27779378 DOI: 10.1021/acschembio.6b00706] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The ever increasing bioactivity data that are produced nowadays allow exhaustive data mining and knowledge discovery approaches that change chemical biology research. A wealth of chemoinformatics tools, web services, and applications therefore exists that supports a careful evaluation and analysis of experimental data to draw conclusions that can influence the further development of chemical probes and potential lead structures. This review focuses on open-source approaches that can be handled by scientists who are not familiar with computational methods having no expert knowledge in chemoinformatics and modeling. Our aim is to present an easily manageable toolbox for support of every day laboratory work. This includes, among other things, the available bioactivity and related molecule databases as well as tools to handle and analyze in-house data.
Collapse
Affiliation(s)
- Lina Humbeck
- Faculty
of Chemistry and
Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| | - Oliver Koch
- Faculty
of Chemistry and
Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| |
Collapse
|
47
|
Vilar S, Quezada E, Uriarte E, Costanzi S, Borges F, Viña D, Hripcsak G. Computational Drug Target Screening through Protein Interaction Profiles. Sci Rep 2016; 6:36969. [PMID: 27845365 PMCID: PMC5109486 DOI: 10.1038/srep36969] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/24/2016] [Indexed: 11/11/2022] Open
Abstract
The development of computational methods to discover novel drug-target interactions on a large scale is of great interest. We propose a new method for virtual screening based on protein interaction profile similarity to discover new targets for molecules, including existing drugs. We calculated Target Interaction Profile Fingerprints (TIPFs) based on ChEMBL database to evaluate drug similarity and generated new putative compound-target candidates from the non-intersecting targets in each pair of compounds. A set of drugs was further studied in monoamine oxidase B (MAO-B) and cyclooxygenase-1 (COX-1) enzyme through molecular docking and experimental assays. The drug ethoxzolamide and the natural compound piperlongumine, present in Piper longum L, showed hMAO-B activity with IC50 values of 25 and 65 μM respectively. Five candidates, including lapatinib, SB-202190, RO-316233, GW786460X and indirubin-3′-monoxime were tested against human COX-1. Compounds SB-202190 and RO-316233 showed a IC50 in hCOX-1 of 24 and 25 μM respectively (similar range as potent inhibitors such as diclofenac and indomethacin in the same experimental conditions). Lapatinib and indirubin-3′-monoxime showed moderate hCOX-1 activity (19.5% and 28% of enzyme inhibition at 25 μM respectively). Our modeling constitutes a multi-target predictor for large scale virtual screening with potential in lead discovery, repositioning and drug safety.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA.,Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Elías Quezada
- CIQUP, Department of Chemistry &Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Eugenio Uriarte
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Stefano Costanzi
- Department of Chemistry, American University, 20016 Washington, DC, USA
| | - Fernanda Borges
- CIQUP, Department of Chemistry &Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Dolores Viña
- Department of Pharmacology, CIMUS, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
48
|
Martínez-Jiménez F, Marti-Renom MA. Should network biology be used for drug discovery? Expert Opin Drug Discov 2016; 11:1135-1137. [PMID: 27635856 DOI: 10.1080/17460441.2016.1236786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Francisco Martínez-Jiménez
- a CNAG-CRG, Centre for Genomic Regulation (CRG) , Barcelona Institute of Science and Technology (BIST) , Barcelona , Spain.,b Gene Regulation, Stem Cells and Cancer Program , Centre for Genomic Regulation (CRG) , Barcelona , Spain.,c Universitat Pompeu Fabra (UPF) , Barcelona , Spain
| | - Marc A Marti-Renom
- a CNAG-CRG, Centre for Genomic Regulation (CRG) , Barcelona Institute of Science and Technology (BIST) , Barcelona , Spain.,b Gene Regulation, Stem Cells and Cancer Program , Centre for Genomic Regulation (CRG) , Barcelona , Spain.,c Universitat Pompeu Fabra (UPF) , Barcelona , Spain.,d ICREA, Pg. Lluís Companys , Barcelona , Spain
| |
Collapse
|
49
|
Siragusa L, Luciani R, Borsari C, Ferrari S, Costi MP, Cruciani G, Spyrakis F. Comparing Drug Images and Repurposing Drugs with BioGPS and FLAPdock: The Thymidylate Synthase Case. ChemMedChem 2016; 11:1653-66. [PMID: 27404817 DOI: 10.1002/cmdc.201600121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/08/2016] [Indexed: 12/14/2022]
Abstract
Repurposing and repositioning drugs has become a frequently pursued and successful strategy in the current era, as new chemical entities are increasingly difficult to find and get approved. Herein we report an integrated BioGPS/FLAPdock pipeline for rapid and effective off-target identification and drug repurposing. Our method is based on the structural and chemical properties of protein binding sites, that is, the ligand image, encoded in the GRID molecular interaction fields (MIFs). Protein similarity is disclosed through the BioGPS algorithm by measuring the pockets' overlap according to which pockets are clustered. Co-crystallized and known ligands can be cross-docked among similar targets, selected for subsequent in vitro binding experiments, and possibly improved for inhibitory potency. We used human thymidylate synthase (TS) as a test case and searched the entire RCSB Protein Data Bank (PDB) for similar target pockets. We chose casein kinase IIα as a control and tested a series of its inhibitors against the TS template. Ellagic acid and apigenin were identified as TS inhibitors, and various flavonoids were selected and synthesized in a second-round selection. The compounds were demonstrated to be active in the low-micromolar range.
Collapse
Affiliation(s)
- Lydia Siragusa
- Molecular Discovery Limited, 215 Marsh Road, Pinner Middlesex, London, HA5 5NE, UK
| | - Rosaria Luciani
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Chiara Borsari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Stefania Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123, Perugia, Italy
| | - Francesca Spyrakis
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy. .,Department of Food Science, University of Parma, Viale delle Scienze 17A, 43124, Parma, Italy.
| |
Collapse
|
50
|
Hsin KY, Matsuoka Y, Asai Y, Kamiyoshi K, Watanabe T, Kawaoka Y, Kitano H. systemsDock: a web server for network pharmacology-based prediction and analysis. Nucleic Acids Res 2016; 44:W507-13. [PMID: 27131384 PMCID: PMC4987901 DOI: 10.1093/nar/gkw335] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/08/2016] [Accepted: 04/15/2016] [Indexed: 11/14/2022] Open
Abstract
We present systemsDock, a web server for network pharmacology-based prediction and analysis, which permits docking simulation and molecular pathway map for comprehensive characterization of ligand selectivity and interpretation of ligand action on a complex molecular network. It incorporates an elaborately designed scoring function for molecular docking to assess protein-ligand binding potential. For large-scale screening and ease of investigation, systemsDock has a user-friendly GUI interface for molecule preparation, parameter specification and result inspection. Ligand binding potentials against individual proteins can be directly displayed on an uploaded molecular interaction map, allowing users to systemically investigate network-dependent effects of a drug or drug candidate. A case study is given to demonstrate how systemsDock can be used to discover a test compound's multi-target activity. systemsDock is freely accessible at http://systemsdock.unit.oist.jp/.
Collapse
Affiliation(s)
- Kun-Yi Hsin
- Integrated Open Systems Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0412, Japan
| | - Yukiko Matsuoka
- The Systems Biology Institute, Minato, Tokyo 108-0071, Japan
| | - Yoshiyuki Asai
- Integrated Open Systems Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0412, Japan
| | - Kyota Kamiyoshi
- Integrated Open Systems Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0412, Japan
| | - Tokiko Watanabe
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53711, USA
| | - Hiroaki Kitano
- Integrated Open Systems Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0412, Japan The Systems Biology Institute, Minato, Tokyo 108-0071, Japan Laboratory for Disease Systems Modeling, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| |
Collapse
|