1
|
Li Y, Wang S, Zhang Y, Ren C, Liu T, Liu Y, Pang S. IRPCA: An Interpretable Robust Principal Component Analysis Framework for Inferring miRNA-Drug Associations. J Chem Inf Model 2025; 65:2432-2442. [PMID: 39980166 DOI: 10.1021/acs.jcim.4c02385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Recent evidence indicates that microribonucleic acids (miRNAs) are crucial in modulating drug sensitivity by orchestrating the expression of genes involved in drug metabolism and its pharmacological effects. Existing predictive methods struggle to extract features related to miRNAs and drugs, often overlooking the significance of data noise and the limitations of using a single similarity measure. To address these limitations, we propose an interpretable robust principal component analysis framework (IRPCA). IRPCA enhances the robustness of the model by employing a nonconvex low-rank approximation, thereby offering greater flexibility. Interpretability is ensured by analyzing low-rank matrix decomposition, which clarifies how miRNAs interact with drugs. Gaussian interaction profile kernel (GIPK) similarities are introduced to compute integrated similarities between miRNAs and drugs, addressing the issue of the single similarity feature. IRPCA is subsequently utilized to extract pertinent features, and a fully connected neural network is employed to generate the ultimate prediction scores. To assess the efficacy of IRPCA, we implemented 5-fold cross-validation (CV), which outperformed other leading methods, achieving the highest area under the curve (AUC) value of 0.9653. Additionally, case studies provide additional evidence supporting the efficacy of IRPCA.
Collapse
Affiliation(s)
- Yunyin Li
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Shudong Wang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum (East China), Qingdao 266580, China
- State Key Laboratory of Chemical Safety, China University of Petroleum (East China), Qingdao 266580, China
- Shandong Key Laboratory of Intelligent Oil and Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao 266525, China
| | - Chuanru Ren
- School of Computer Science and Technology, Tongji University, Shanghai 201804, China
| | - Tiyao Liu
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Yingye Liu
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Shanchen Pang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum (East China), Qingdao 266580, China
- State Key Laboratory of Chemical Safety, China University of Petroleum (East China), Qingdao 266580, China
- Shandong Key Laboratory of Intelligent Oil and Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
2
|
Galeano D, Imrat, Haltom J, Andolino C, Yousey A, Zaksas V, Das S, Baylin SB, Wallace DC, Slack FJ, Enguita FJ, Wurtele ES, Teegarden D, Meller R, Cifuentes D, Beheshti A. sChemNET: a deep learning framework for predicting small molecules targeting microRNA function. Nat Commun 2024; 15:9149. [PMID: 39443444 PMCID: PMC11500171 DOI: 10.1038/s41467-024-49813-w] [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: 08/18/2023] [Accepted: 06/14/2024] [Indexed: 10/25/2024] Open
Abstract
MicroRNAs (miRNAs) have been implicated in human disorders, from cancers to infectious diseases. Targeting miRNAs or their target genes with small molecules offers opportunities to modulate dysregulated cellular processes linked to diseases. Yet, predicting small molecules associated with miRNAs remains challenging due to the small size of small molecule-miRNA datasets. Herein, we develop a generalized deep learning framework, sChemNET, for predicting small molecules affecting miRNA bioactivity based on chemical structure and sequence information. sChemNET overcomes the limitation of sparse chemical information by an objective function that allows the neural network to learn chemical space from a large body of chemical structures yet unknown to affect miRNAs. We experimentally validated small molecules predicted to act on miR-451 or its targets and tested their role in erythrocyte maturation during zebrafish embryogenesis. We also tested small molecules targeting the miR-181 network and other miRNAs using in-vitro and in-vivo experiments. We demonstrate that our machine-learning framework can predict bioactive small molecules targeting miRNAs or their targets in humans and other mammalian organisms.
Collapse
Affiliation(s)
- Diego Galeano
- Department of Electronics and Mechatronics Engineering, Facultad de Ingeniería, Universidad Nacional de Asunción - FIUNA, Luque, Paraguay.
- COVID-19 International Research Team, Medford, MA, USA.
| | - Imrat
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jeffrey Haltom
- COVID-19 International Research Team, Medford, MA, USA
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chaylen Andolino
- Department of Nutrition Science, Purdue University, Indiana, USA
- Purdue Institute for Cancer Research, Purdue University, Indiana, USA
| | - Aliza Yousey
- COVID-19 International Research Team, Medford, MA, USA
- Neuroscience Institute, Department of Neurobiology/ Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Victoria Zaksas
- COVID-19 International Research Team, Medford, MA, USA
- Center for Translational Data Science, University of Chicago, Chicago, IL, USA
- Clever Research Lab, Springfield, IL, USA
| | - Saswati Das
- COVID-19 International Research Team, Medford, MA, USA
- Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Stephen B Baylin
- COVID-19 International Research Team, Medford, MA, USA
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Van Andel Institute, Grand Rapids, MI, USA
| | - Douglas C Wallace
- COVID-19 International Research Team, Medford, MA, USA
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frank J Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Francisco J Enguita
- COVID-19 International Research Team, Medford, MA, USA
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Eve Syrkin Wurtele
- Bioinformatics and Computational Biology Program, Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA
| | - Dorothy Teegarden
- Department of Nutrition Science, Purdue University, Indiana, USA
- Purdue Institute for Cancer Research, Purdue University, Indiana, USA
| | - Robert Meller
- COVID-19 International Research Team, Medford, MA, USA
- Neuroscience Institute, Department of Neurobiology/ Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Daniel Cifuentes
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Afshin Beheshti
- COVID-19 International Research Team, Medford, MA, USA
- Blue Marble Space Institute of Science, NASA Ames Research Center, Moffett Field, CA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Jurj A, Fontana B, Varani G, Calin GA. Small molecules targeting microRNAs: new opportunities and challenges in precision cancer therapy. Trends Cancer 2024; 10:809-824. [PMID: 39107162 PMCID: PMC11961049 DOI: 10.1016/j.trecan.2024.06.006] [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: 11/27/2023] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 08/09/2024]
Abstract
Noncoding RNAs, especially miRNAs, play a pivotal role in cancer initiation and metastasis, underscoring their susceptibility to precise modulation via small molecule inhibitors. This review examines the innovative strategy of targeting oncogenic miRNAs with small drug-like molecules, an approach that can reshape the cancer treatment landscape. We review the current understanding of the multifaceted roles of miRNAs in oncogenesis, highlighting emerging therapeutic paradigms that have the potential to expand cancer treatment options. As research on small molecule inhibitors of miRNA is still in its early stages, ongoing investigative efforts and the development of new technologies and chemical matter are essential to fulfill the significant potential of this innovative approach to cancer treatment.
Collapse
Affiliation(s)
- Ancuta Jurj
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beatrice Fontana
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Gabriele Varani
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA.
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Sun XY, Hou ZJ, Zhang WG, Chen Y, Yao HB. HTFSMMA: Higher-Order Topological Guided Small Molecule-MicroRNA Associations Prediction. J Comput Biol 2024; 31:886-906. [PMID: 39109562 DOI: 10.1089/cmb.2024.0587] [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] [Indexed: 09/10/2024] Open
Abstract
Small molecules (SMs) play a pivotal role in regulating microRNAs (miRNAs). Existing prediction methods for associations between SM-miRNA have overlooked crucial aspects: the incorporation of local topological features between nodes, which represent either SMs or miRNAs, and the effective fusion of node features with topological features. This study introduces a novel approach, termed high-order topological features for SM-miRNA association prediction (HTFSMMA), which specifically addresses these limitations. Initially, an association graph is formed by integrating SM-miRNA association data, SM similarity, and miRNA similarity. Subsequently, we focus on the local information of links and propose target neighborhood graph convolutional network for extracting local topological features. Then, HTFSMMA employs graph attention networks to amalgamate these local features, thereby establishing a platform for the acquisition of high-order features through random walks. Finally, the extracted features are integrated into the multilayer perceptron to derive the association prediction scores. To demonstrate the performance of HTFSMMA, we conducted comprehensive evaluations including five-fold cross-validation, leave-one-out cross-validation (LOOCV), SM-fixed local LOOCV, and miRNA-fixed local LOOCV. The area under receiver operating characteristic curve values were 0.9958 ± 0.0024 (0.8722 ± 0.0021), 0.9986 (0.9504), 0.9974 (0.9111), and 0.9977 (0.9074), respectively. Our findings demonstrate the superior performance of HTFSMMA over existing approaches. In addition, three case studies and the DeLong test have confirmed the effectiveness of the proposed method. These results collectively underscore the significance of HTFSMMA in facilitating the inference of associations between SMs and miRNAs.
Collapse
Affiliation(s)
- Xiao-Yan Sun
- School of Computer Science and Artificial Intelligence & Aliyun Big Data, Changzhou University, Changzhou, China
| | - Zhen-Jie Hou
- School of Computer Science and Artificial Intelligence & Aliyun Big Data, Changzhou University, Changzhou, China
| | - Wen-Guang Zhang
- School of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Yan Chen
- School of Computer Science and Artificial Intelligence & Aliyun Big Data, Changzhou University, Changzhou, China
| | - Hai-Bin Yao
- School of Computer Science and Artificial Intelligence & Aliyun Big Data, Changzhou University, Changzhou, China
| |
Collapse
|
5
|
Wang S, Liu T, Ren C, Zhao Y, Qiao S, Zhang Y, Pang S. Heterogeneous graph inference with range constrainted L 2,1-collaborative matrix factorization for small molecule-miRNA association prediction. Comput Biol Chem 2024; 110:108078. [PMID: 38677013 DOI: 10.1016/j.compbiolchem.2024.108078] [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: 02/06/2024] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024]
Abstract
MicroRNAs (miRNAs) play a vital role in regulating gene expression and various biological processes. As a result, they have been identified as effective targets for small molecule (SM) drugs in disease treatment. Heterogeneous graph inference stands as a classical approach for predicting SM-miRNA associations, showcasing commendable convergence accuracy and speed. However, most existing methods do not adequately address the inherent sparsity in SM-miRNA association networks, and imprecise SM/miRNA similarity metrics reduce the accuracy of predicting SM-miRNA associations. In this research, we proposed a heterogeneous graph inference with range constrained L2,1-collaborative matrix factorization (HGIRCLMF) method to predict potential SM-miRNA associations. First, we computed the multi-source similarities of SM/miRNA and integrated these similarity information into a comprehensive SM/miRNA similarity. This step improved the accuracy of SM and miRNA similarity, ensuring reliability for the subsequent inference of the heterogeneity map. Second, we used a range constrained L2,1-collaborative matrix factorization (RCLMF) model to pre-populate the SM-miRNA association matrix with missing values. In this step, we developed a novel matrix decomposition method that enhances the robustness and formative nature of SM-miRNA edges between SM networks and miRNA networks. Next, we built a well-established SM-miRNA heterogeneous network utilizing the processed biological information. Finally, HGIRCLMF used this network data to infer unknown association pair scores. We implemented four cross-validation experiments on two distinct datasets, and HGIRCLMF acquired the highest areas under the curve, surpassing six state-of-the-art computational approaches. Furthermore, we performed three case studies to validate the predictive power of our method in practical application.
Collapse
Affiliation(s)
- Shudong Wang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Tiyao Liu
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Chuanru Ren
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Yawu Zhao
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Sibo Qiao
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Yuanyuan Zhang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao 266525, China.
| | - Shanchen Pang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| |
Collapse
|
6
|
Guan YJ, Yu CQ, Li LP, You ZH, Wei MM, Wang XF, Yang C, Guo LX. MHESMMR: a multilevel model for predicting the regulation of miRNAs expression by small molecules. BMC Bioinformatics 2024; 25:6. [PMID: 38166644 PMCID: PMC10763044 DOI: 10.1186/s12859-023-05629-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
According to the expression of miRNA in pathological processes, miRNAs can be divided into oncogenes or tumor suppressors. Prediction of the regulation relations between miRNAs and small molecules (SMs) becomes a vital goal for miRNA-target therapy. But traditional biological approaches are laborious and expensive. Thus, there is an urgent need to develop a computational model. In this study, we proposed a computational model to predict whether the regulatory relationship between miRNAs and SMs is up-regulated or down-regulated. Specifically, we first use the Large-scale Information Network Embedding (LINE) algorithm to construct the node features from the self-similarity networks, then use the General Attributed Multiplex Heterogeneous Network Embedding (GATNE) algorithm to extract the topological information from the attribute network, and finally utilize the Light Gradient Boosting Machine (LightGBM) algorithm to predict the regulatory relationship between miRNAs and SMs. In the fivefold cross-validation experiment, the average accuracies of the proposed model on the SM2miR dataset reached 79.59% and 80.37% for up-regulation pairs and down-regulation pairs, respectively. In addition, we compared our model with another published model. Moreover, in the case study for 5-FU, 7 of 10 candidate miRNAs are confirmed by related literature. Therefore, we believe that our model can promote the research of miRNA-targeted therapy.
Collapse
Affiliation(s)
- Yong-Jian Guan
- School of Information Engineering, Xijing University, Xi'an, China
| | - Chang-Qing Yu
- School of Information Engineering, Xijing University, Xi'an, China.
| | - Li-Ping Li
- College of Grassland and Environment Sciences, Xinjiang Agricultural University, Urumqi, China.
- College of Agriculture and Forestry, Longdong University, Qingyang, China.
| | - Zhu-Hong You
- School of Computer Science, North-Western Polytechnical University, Xi'an, China
| | - Meng-Meng Wei
- School of Information Engineering, Xijing University, Xi'an, China
| | - Xin-Fei Wang
- School of Information Engineering, Xijing University, Xi'an, China
| | - Chen Yang
- School of Information Engineering, Xijing University, Xi'an, China
| | - Lu-Xiang Guo
- School of Information Engineering, Xijing University, Xi'an, China
| |
Collapse
|
7
|
Vasu S, Saracino G, Darden CM, Kumano K, Liu Y, Lawrence MC, Naziruddin B. Clinical and biological significance of circulating miRNAs in chronic pancreatitis patients undergoing total pancreatectomy with islet autotransplantation. Clin Transl Med 2023; 13:e1434. [PMID: 37846205 PMCID: PMC10579997 DOI: 10.1002/ctm2.1434] [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: 03/27/2023] [Revised: 09/12/2023] [Accepted: 09/30/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Specific microRNAs (miRNAs) were elevated in chronic pancreatitis (CP) patients during islet infusion after total pancreatectomy (TPIAT). We aimed to identify circulating miRNA signatures of pancreatic damage, predict miRNA-mRNA networks to identify potential links to CP pathogenesis and identify islet isolation and transplantation functional outcomes. METHODS Small RNA sequencing was performed to identify distinct circulating miRNA signatures in CP. Plasma miRNAs were measured using miRCURY LNA SYBR green quantitative real-time polymerase chain reaction assays. Correlation analyses were performed using R software. The miRNA target and disease interactions were determined using miRNet and the miRNA enrichment and annotation tool. RESULTS Alterations were found in circulating miRNAs in CP patients compared to healthy controls. Further studies were conducted on 12 circulating miRNAs enriched in the pancreas, other tissues and other diseases including cancer and fibrosis. Approximately 2888 mRNAs in the pancreas were their targets, demonstrating interactions with 76 small molecules. Three miRNAs exhibited interactions with morphine and five exhibited interactions with glucose. The miRNA panel targeted 22 genes associated with pancreatitis. The islet-specific, acinar cell-specific and liver-specific miRNAs were elevated at 6 h after islet infusion and returned to baseline levels 3 months after TPIAT. Circulating levels of miRNAs returned to pre-transplant levels 1-year post-transplant. Circulating miRNAs measured before and 6 h after islet infusion were directly or inversely associated with metabolic outcomes at 3 and 6 months post-transplant. CONCLUSIONS miRNAs may contribute to CP pathogenesis, and elevated circulating levels may be specific to pancreatic inflammation and fibrosis, warranting further investigation.
Collapse
Affiliation(s)
- Srividya Vasu
- Islet Cell LaboratoryBaylor Scott and White Research InstituteDallasTexasUSA
| | - Giovanna Saracino
- Baylor Simmons Transplant InstituteBaylor University Medical CenterDallasTexasUSA
| | - Carly M. Darden
- Islet Cell LaboratoryBaylor Scott and White Research InstituteDallasTexasUSA
| | - Kenjiro Kumano
- Department of Gastroenterological SurgeryOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Yang Liu
- The University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Michael C. Lawrence
- Islet Cell LaboratoryBaylor Scott and White Research InstituteDallasTexasUSA
| | - Bashoo Naziruddin
- Baylor Simmons Transplant InstituteBaylor University Medical CenterDallasTexasUSA
| |
Collapse
|
8
|
Sun J, Xu M, Ru J, James-Bott A, Xiong D, Wang X, Cribbs AP. Small molecule-mediated targeting of microRNAs for drug discovery: Experiments, computational techniques, and disease implications. Eur J Med Chem 2023; 257:115500. [PMID: 37262996 PMCID: PMC11554572 DOI: 10.1016/j.ejmech.2023.115500] [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: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Small molecules have been providing medical breakthroughs for human diseases for more than a century. Recently, identifying small molecule inhibitors that target microRNAs (miRNAs) has gained importance, despite the challenges posed by labour-intensive screening experiments and the significant efforts required for medicinal chemistry optimization. Numerous experimentally-verified cases have demonstrated the potential of miRNA-targeted small molecule inhibitors for disease treatment. This new approach is grounded in their posttranscriptional regulation of the expression of disease-associated genes. Reversing dysregulated gene expression using this mechanism may help control dysfunctional pathways. Furthermore, the ongoing improvement of algorithms has allowed for the integration of computational strategies built on top of laboratory-based data, facilitating a more precise and rational design and discovery of lead compounds. To complement the use of extensive pharmacogenomics data in prioritising potential drugs, our previous work introduced a computational approach based on only molecular sequences. Moreover, various computational tools for predicting molecular interactions in biological networks using similarity-based inference techniques have been accumulated in established studies. However, there are a limited number of comprehensive reviews covering both computational and experimental drug discovery processes. In this review, we outline a cohesive overview of both biological and computational applications in miRNA-targeted drug discovery, along with their disease implications and clinical significance. Finally, utilizing drug-target interaction (DTIs) data from DrugBank, we showcase the effectiveness of deep learning for obtaining the physicochemical characterization of DTIs.
Collapse
Affiliation(s)
- Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| | - Miaoer Xu
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Jinlong Ru
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, 85354, Germany
| | - Anna James-Bott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
9
|
Qu J, Song Z, Cheng X, Jiang Z, Zhou J. Neighborhood-based inference and restricted Boltzmann machine for small molecule-miRNA associations prediction. PeerJ 2023; 11:e15889. [PMID: 37641598 PMCID: PMC10460564 DOI: 10.7717/peerj.15889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
Background A growing number of experiments have shown that microRNAs (miRNAs) can be used as target of small molecules (SMs) to regulate gene expression for treating diseases. Therefore, identifying SM-related miRNAs is helpful for the treatment of diseases in the domain of medical investigation. Methods This article presents a new computational model, called NIRBMSMMA (neighborhood-based inference (NI) and restricted Boltzmann machine (RBM)), which we developed to identify potential small molecule-miRNA associations (NIRBMSMMA). First, grounded on known SM-miRNAs associations, SM similarity and miRNA similarity, NI was used to predict score of an unknown SM-miRNA pair by reckoning the sum of known associations between neighbors of the SM (miRNA) and the miRNA (SM). Second, utilizing a two-layered generative stochastic artificial neural network, RBM was used to predict SM-miRNA association by learning potential probability distribution from known SM-miRNA associations. At last, an ensemble learning model was conducted to combine NI and RBM for identifying potential SM-miRNA associations. Results Furthermore, we conducted global leave one out cross validation (LOOCV), miRNA-fixed LOOCV, SM-fixed LOOCV and five-fold cross validation to assess performance of NIRBMSMMA based on three datasets. Results showed that NIRBMSMMA obtained areas under the curve (AUC) of 0.9912, 0.9875, 0.8376 and 0.9898 ± 0.0009 under global LOOCV, miRNA-fixed LOOCV, SM-fixed LOOCV and five-fold cross validation based on dataset 1, respectively. For dataset 2, the AUCs are 0.8645, 0.8720, 0.7066 and 0.8547 ± 0.0046 in turn. For dataset 3, the AUCs are 0.9884, 0.9802, 0.8239 and 0.9870 ± 0.0015 in turn. Also, we conducted case studies to further assess the predictive performance of NIRBMSMMA. These results illustrated the proposed model is a useful tool in predicting potential SM-miRNA associations.
Collapse
Affiliation(s)
- Jia Qu
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, Jiangsu, China
| | - Zihao Song
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, Jiangsu, China
| | - Xiaolong Cheng
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, Jiangsu, China
| | - Zhibin Jiang
- Department of Computer Science and Engineering, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jie Zhou
- Department of Computer Science and Engineering, Shaoxing University, Shaoxing, Zhejiang, China
| |
Collapse
|
10
|
Wang S, Ren C, Zhang Y, Pang S, Qiao S, Wu W, Lin B. AMCSMMA: Predicting Small Molecule-miRNA Potential Associations Based on Accurate Matrix Completion. Cells 2023; 12:cells12081123. [PMID: 37190032 DOI: 10.3390/cells12081123] [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: 02/08/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Exploring potential associations between small molecule drugs (SMs) and microRNAs (miRNAs) is significant for drug development and disease treatment. Since biological experiments are expensive and time-consuming, we propose a computational model based on accurate matrix completion for predicting potential SM-miRNA associations (AMCSMMA). Initially, a heterogeneous SM-miRNA network is constructed, and its adjacency matrix is taken as the target matrix. An optimization framework is then proposed to recover the target matrix with the missing values by minimizing its truncated nuclear norm, an accurate, robust, and efficient approximation to the rank function. Finally, we design an effective two-step iterative algorithm to solve the optimization problem and obtain the prediction scores. After determining the optimal parameters, we conduct four kinds of cross-validation experiments based on two datasets, and the results demonstrate that AMCSMMA is superior to the state-of-the-art methods. In addition, we implement another validation experiment, in which more evaluation metrics in addition to the AUC are introduced and finally achieve great results. In two types of case studies, a large number of SM-miRNA pairs with high predictive scores are confirmed by the published experimental literature. In summary, AMCSMMA has superior performance in predicting potential SM-miRNA associations, which can provide guidance for biological experiments and accelerate the discovery of new SM-miRNA associations.
Collapse
Affiliation(s)
- Shudong Wang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Chuanru Ren
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Yulin Zhang
- College of Mathematics and Systems Science, Shandong University of Science and Technology, Qingdao 266580, China
| | - Shanchen Pang
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Sibo Qiao
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Wenhao Wu
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| | - Boyang Lin
- College of Computer Science and Technology, Qingdao Institute of Software, China University of Petroleum, Qingdao 266580, China
| |
Collapse
|
11
|
Wang MN, Li Y, Lei LL, Ding DW, Xie XJ. Combining non-negative matrix factorization with graph Laplacian regularization for predicting drug-miRNA associations based on multi-source information fusion. Front Pharmacol 2023; 14:1132012. [PMID: 36817132 PMCID: PMC9931722 DOI: 10.3389/fphar.2023.1132012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Increasing evidences suggest that miRNAs play a key role in the occurrence and progression of many complex human diseases. Therefore, targeting dysregulated miRNAs with small molecule drugs in the clinical has become a new treatment. Nevertheless, it is high cost and time-consuming for identifying miRNAs-targeted with drugs by biological experiments. Thus, more reliable computational method for identification associations of drugs with miRNAs urgently need to be developed. In this study, we proposed an efficient method, called GNMFDMA, to predict potential associations of drug with miRNA by combining graph Laplacian regularization with non-negative matrix factorization. We first calculated the overall similarity matrices of drugs and miRNAs according to the collected different biological information. Subsequently, the new drug-miRNA association adjacency matrix was reformulated based on the K nearest neighbor profiles so as to put right the false negative associations. Finally, graph Laplacian regularization collaborative non-negative matrix factorization was used to calculate the association scores of drugs with miRNAs. In the cross validation, GNMFDMA obtains AUC of 0.9193, which outperformed the existing methods. In addition, case studies on three common drugs (i.e., 5-Aza-CdR, 5-FU and Gemcitabine), 30, 31 and 34 of the top-50 associations inferred by GNMFDMA were verified. These results reveal that GNMFDMA is a reliable and efficient computational approach for identifying the potential drug-miRNA associations.
Collapse
Affiliation(s)
- Mei-Neng Wang
- School of Mathematics and Computer Science, Yichun University, Yichun, China
| | - Yu Li
- School of Information Engineering, Inner Mongolia University of Science and Technology, Baotou, China,*Correspondence: Yu Li,
| | - Li-Lan Lei
- School of Mathematics and Computer Science, Yichun University, Yichun, China
| | - De-Wu Ding
- School of Mathematics and Computer Science, Yichun University, Yichun, China
| | - Xue-Jun Xie
- School of Mathematics and Computer Science, Yichun University, Yichun, China
| |
Collapse
|
12
|
Sun J, Ru J, Ramos-Mucci L, Qi F, Chen Z, Chen S, Cribbs AP, Deng L, Wang X. DeepsmirUD: Prediction of Regulatory Effects on microRNA Expression Mediated by Small Molecules Using Deep Learning. Int J Mol Sci 2023; 24:1878. [PMID: 36768205 PMCID: PMC9915273 DOI: 10.3390/ijms24031878] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/26/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Aberrant miRNA expression has been associated with a large number of human diseases. Therefore, targeting miRNAs to regulate their expression levels has become an important therapy against diseases that stem from the dysfunction of pathways regulated by miRNAs. In recent years, small molecules have demonstrated enormous potential as drugs to regulate miRNA expression (i.e., SM-miR). A clear understanding of the mechanism of action of small molecules on the upregulation and downregulation of miRNA expression allows precise diagnosis and treatment of oncogenic pathways. However, outside of a slow and costly process of experimental determination, computational strategies to assist this on an ad hoc basis have yet to be formulated. In this work, we developed, to the best of our knowledge, the first cross-platform prediction tool, DeepsmirUD, to infer small-molecule-mediated regulatory effects on miRNA expression (i.e., upregulation or downregulation). This method is powered by 12 cutting-edge deep-learning frameworks and achieved AUC values of 0.843/0.984 and AUCPR values of 0.866/0.992 on two independent test datasets. With a complementarily constructed network inference approach based on similarity, we report a significantly improved accuracy of 0.813 in determining the regulatory effects of nearly 650 associated SM-miR relations, each formed with either novel small molecule or novel miRNA. By further integrating miRNA-cancer relationships, we established a database of potential pharmaceutical drugs from 1343 small molecules for 107 cancer diseases to understand the drug mechanisms of action and offer novel insight into drug repositioning. Furthermore, we have employed DeepsmirUD to predict the regulatory effects of a large number of high-confidence associated SM-miR relations. Taken together, our method shows promise to accelerate the development of potential miRNA targets and small molecule drugs.
Collapse
Affiliation(s)
- Jianfeng Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Jinlong Ru
- Institute of Virology, Helmholtz Centre Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Lorenzo Ramos-Mucci
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Fei Qi
- Institute of Genomics, School of Medicine, Huaqiao University, Xiamen 362021, China
| | - Zihao Chen
- Department of Computational Biology for Drug Discovery, Biolife Biotechnology Ltd., Zhumadian 463200, China
| | - Suyuan Chen
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., Otto-Hahn-Str asse 6b, 44227 Dortmund, Germany
| | - Adam P. Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Li Deng
- Institute of Virology, Helmholtz Centre Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
13
|
Xiao H, Yang X, Zhang Y, Zhang Z, Zhang G, Zhang BT. RNA-targeted small-molecule drug discoveries: a machine-learning perspective. RNA Biol 2023; 20:384-397. [PMID: 37337437 PMCID: PMC10283424 DOI: 10.1080/15476286.2023.2223498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
In the past two decades, machine learning (ML) has been extensively adopted in protein-targeted small molecule (SM) discovery. Once trained, ML models could exert their predicting abilities on large volumes of molecules within a short time. However, applying ML approaches to discover RNA-targeted SMs is still in its early stages. This is primarily because of the intrinsic structural instability of RNA molecules that impede the structure-based screening or designing of RNA-targeted SMs. Recently, with more studies revealing RNA structures and a growing number of RNA-targeted ligands being identified, it resulted in an increased interest in the field of drugging RNA. Undeniably, intracellular RNA is much more abundant than protein and, if successfully targeted, will be a major alternative target for therapeutics. Therefore, in this context, as well as under the premise of having RNA-related research data, ML-based methods can get involved in improving the speed of traditional experimental processes. [Figure: see text].
Collapse
Affiliation(s)
- Huan Xiao
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yihao Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
14
|
MFIDMA: A Multiple Information Integration Model for the Prediction of Drug-miRNA Associations. BIOLOGY 2022; 12:biology12010041. [PMID: 36671734 PMCID: PMC9855084 DOI: 10.3390/biology12010041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Abnormal microRNA (miRNA) functions play significant roles in various pathological processes. Thus, predicting drug-miRNA associations (DMA) may hold great promise for identifying the potential targets of drugs. However, discovering the associations between drugs and miRNAs through wet experiments is time-consuming and laborious. Therefore, it is significant to develop computational prediction methods to improve the efficiency of identifying DMA on a large scale. In this paper, a multiple features integration model (MFIDMA) is proposed to predict drug-miRNA association. Specifically, we first formulated known DMA as a bipartite graph and utilized structural deep network embedding (SDNE) to learn the topological features from the graph. Second, the Word2vec algorithm was utilized to construct the attribute features of the miRNAs and drugs. Third, two kinds of features were entered into the convolution neural network (CNN) and deep neural network (DNN) to integrate features and predict potential target miRNAs for the drugs. To evaluate the MFIDMA model, it was implemented on three different datasets under a five-fold cross-validation and achieved average AUCs of 0.9407, 0.9444 and 0.8919. In addition, the MFIDMA model showed reliable results in the case studies of Verapamil and hsa-let-7c-5p, confirming that the proposed model can also predict DMA in real-world situations. The model was effective in analyzing the neighbors and topological features of the drug-miRNA network by SDNE. The experimental results indicated that the MFIDMA is an accurate and robust model for predicting potential DMA, which is significant for miRNA therapeutics research and drug discovery.
Collapse
|
15
|
Li J, Lin H, Wang Y, Li Z, Wu B. Prediction of potential small molecule-miRNA associations based on heterogeneous network representation learning. Front Genet 2022; 13:1079053. [PMID: 36531225 PMCID: PMC9755196 DOI: 10.3389/fgene.2022.1079053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2023] Open
Abstract
MicroRNAs (miRNAs) are closely associated with the occurrences and developments of many complex human diseases. Increasing studies have shown that miRNAs emerge as new therapeutic targets of small molecule (SM) drugs. Since traditional experiment methods are expensive and time consuming, it is particularly crucial to find efficient computational approaches to predict potential small molecule-miRNA (SM-miRNA) associations. Considering that integrating multi-source heterogeneous information related with SM-miRNA association prediction would provide a comprehensive insight into the features of both SMs and miRNAs, we proposed a novel model of Small Molecule-MiRNA Association prediction based on Heterogeneous Network Representation Learning (SMMA-HNRL) for more precisely predicting the potential SM-miRNA associations. In SMMA-HNRL, a novel heterogeneous information network was constructed with SM nodes, miRNA nodes and disease nodes. To access and utilize of the topological information of the heterogeneous information network, feature vectors of SM and miRNA nodes were obtained by two different heterogeneous network representation learning algorithms (HeGAN and HIN2Vec) respectively and merged with connect operation. Finally, LightGBM was chosen as the classifier of SMMA-HNRL for predicting potential SM-miRNA associations. The 10-fold cross validations were conducted to evaluate the prediction performance of SMMA-HNRL, it achieved an area under of ROC curve of 0.9875, which was superior to other three state-of-the-art models. With two independent validation datasets, the test experiment results revealed the robustness of our model. Moreover, three case studies were performed. As a result, 35, 37, and 22 miRNAs among the top 50 predicting miRNAs associated with 5-FU, cisplatin, and imatinib were validated by experimental literature works respectively, which confirmed the effectiveness of SMMA-HNRL. The source code and experimental data of SMMA-HNRL are available at https://github.com/SMMA-HNRL/SMMA-HNRL.
Collapse
Affiliation(s)
- Jianwei Li
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
- Hebei Province Key Laboratory of Big Data Calculation, Hebei University of Technology, Tianjin, China
| | - Hongxin Lin
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| | - Yinfei Wang
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| | - Zhiguang Li
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| | - Baoqin Wu
- School of Artificial Intelligence, Institute of Computational Medicine, Hebei University of Technology, Tianjin, China
| |
Collapse
|
16
|
Lee M, Kim PJ, Joe H, Kim HG. Gene-centric multi-omics integration with convolutional encoders for cancer drug response prediction. Comput Biol Med 2022; 151:106192. [PMID: 36327883 DOI: 10.1016/j.compbiomed.2022.106192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/26/2022] [Accepted: 10/08/2022] [Indexed: 12/27/2022]
Abstract
MOTIVATION Tumor heterogeneity, including genetic and transcriptomic characteristics, can reduce the efficacy of anticancer pharmacological therapy, resulting in clinical variability in patient response to therapeutic medications. Multi-omics integration can allow in silico models to provide an additional perspective on a biological system. METHODS In this study, we propose a gene-centric multi-channel (GCMC) architecture to integrate multi-omics for predicting cancer drug response. GCMC transformed multi-omics profiles into a three-dimensional tensor with an additional dimension for omics types. GCMC's convolutional encoders captures multi-omics profiles for each gene and yields gene-centric features to predict drug responses. RESULTS We evaluated GCMC on various datasets, including The Cancer Genome Atlas (TCGA) patients, patient-derived xenografts (PDX) mice models, and the Genomics of Drug Sensitivity in Cancer (GDSC) cell line datasets. GCMC achieved better performance than baseline models, including single-omics models, in more than 75% of 265 drugs from GDSC cell line datasets. Furthermore, as for the clinical applicability of GCMC, it achieved the best performance on TCGA and PDX datasets in terms of both AUPR and AUC. We also analyzed models' capability of integrating multi-omics profiles by measuring the contribution ratio of omics types. GCMC can incorporate multi-omics profiles in various manners to enhance performance for each drug type. These results suggested that GCMC can improve performance and feature extraction capability by integrating multi-omics profiles in a gene-centric manner.
Collapse
Affiliation(s)
- Munhwan Lee
- Biomedical Knowledge Engineering Lab., Seoul National University, 1 Gwanak-ro, Seoul, 08826, Republic of Korea.
| | - Pil-Jong Kim
- Biomedical Knowledge Engineering Lab., Seoul National University, 1 Gwanak-ro, Seoul, 08826, Republic of Korea.
| | - Hyunwhan Joe
- Biomedical Knowledge Engineering Lab., Seoul National University, 1 Gwanak-ro, Seoul, 08826, Republic of Korea.
| | - Hong-Gee Kim
- Biomedical Knowledge Engineering Lab., Seoul National University, 1 Gwanak-ro, Seoul, 08826, Republic of Korea.
| |
Collapse
|
17
|
Wu T, Gao YY, Tang XN, Li Y, Dai J, Zhou S, Wu M, Zhang JJ, Wang SX. Construction of a competing endogenous RNA network to identify drug targets against polycystic ovary syndrome. Hum Reprod 2022; 37:2856-2866. [PMID: 36223608 DOI: 10.1093/humrep/deac218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 08/01/2022] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Would the construction of a competing endogenous RNA (ceRNA) network help identify new drug targets for the development of potential therapies for polycystic ovary syndrome (PCOS)? SUMMARY ANSWER Both Food and Drug Administartion (FDA)-approved and candidate drugs could be identified by combining bioinformatics approaches with clinical sample analysis based on our established ceRNA network. WHAT IS KNOWN ALREADY Thus far, no effective drugs are available for treating PCOS. ceRNAs play crucial roles in multiple diseases, and some of them are in current use as prognostic biomarkers as well as for chemo-response and drug prediction. STUDY DESIGN, SIZE, DURATION For the bioinformatics part, five microarrays of human granulosa cells were considered eligible after applying strict screening criteria and were used to construct the ceRNA network for target identification. For population-based validation, samples from 24 women with and without PCOS were collected from January 2021 to July 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS The public data included 27 unaffected women and 25 women with PCOS, according to the Rotterdam criteria proposed in 2003. The limma and RobustRankAggreg R packages were used to identify differentially expressed messenger RNAs and noncoding RNAs. Gene Ontology, Reactome and Kyoto Encyclopedia of Genes and Gemomes (KEGG) enrichment analyses were performed. A ceRNA network was constructed by integrating the differentially expressed genes and target genes. The population-based validation included human luteinized granulosa cell samples from 12 unaffected women and 12 women with PCOS. Quantitative real-time polymerase chain reaction was conducted to detect the levels of mRNAs and microRNAs (miRNAs). Connectivity map and computational model algorithms were implemented to predict therapeutic drugs from the ceRNA network. Additionally, we compared the predicted drugs with known clinical medications in DrugBank. MAIN RESULTS AND THE ROLE OF CHANCE A set of 10 mRNAs, 11 miRNAs and 53 long non-coding RNAs (lncRNAs) were differentially expressed. Functional enrichment analysis revealed the highest relevance to immune system-related biological processes and signalling pathways, such as cytokine secretion and leucocyte chemotaxis. A ceRNA consisting of two lncRNAs, two miRNAs and five mRNAs was constructed. Through network construction via bioinformatic analysis, we identified some already approved drugs (such as metformin) that could target some molecules in the network as potential drug candidates for PCOS. LARGE SCALE DATA Public sequencing data were obtained from GSE34526, GSE84376, GSE102293, GSE106724 and GSE114419, which have been deposited in the Gene Expression Omnibus database. LIMITATIONS, REASONS FOR CAUTION Experiments, such as immunoprecipitation, luciferase reporter assays and animal model studies, are needed to validate the potential targets in the ceRNA network before the identified drug candidates can be tested using cellular and animal model systems. WIDER IMPLICATIONS OF THE FINDINGS Our findings provide new bioinformatic insight into the possible pathogenesis of PCOS from ceRNA network analysis, which has not been previously studied in the human reproductive field. Our study also reveals some potential drug candidates for the future development of possible therapies against PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the National Key Research and Development Program of China (2021YFC2700400) and the National Natural Science Foundation of China (82001498). The authors have no conflicts of interest to disclose.
Collapse
Affiliation(s)
- Tong Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue-Yue Gao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia-Nan Tang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Dai
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Su Zhou
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Jin Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Xuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Ni J, Cheng X, Ni T, Liang J. Identifying SM-miRNA associations based on layer attention graph convolutional network and matrix decomposition. Front Mol Biosci 2022; 9:1009099. [PMID: 36504714 PMCID: PMC9732030 DOI: 10.3389/fmolb.2022.1009099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/03/2022] [Indexed: 11/27/2022] Open
Abstract
The accurate prediction of potential associations between microRNAs (miRNAs) and small molecule (SM) drugs can enhance our knowledge of how SM cures endogenous miRNA-related diseases. Given that traditional methods for predicting SM-miRNA associations are time-consuming and arduous, a number of computational models have been proposed to anticipate the potential SM-miRNA associations. However, several of these strategies failed to eliminate noise from the known SM-miRNA association information or failed to prioritize the most significant known SM-miRNA associations. Therefore, we proposed a model of Graph Convolutional Network with Layer Attention mechanism for SM-MiRNA Association prediction (GCNLASMMA). Firstly, we obtained the new SM-miRNA associations by matrix decomposition. The new SM-miRNA associations, as well as the integrated SM similarity and miRNA similarity were subsequently incorporated into a heterogeneous network. Finally, a graph convolutional network with an attention mechanism was used to compute the reconstructed SM-miRNA association matrix. Furthermore, four types of cross validations and two types of case studies were performed to assess the performance of GCNLASMMA. In cross validation, global Leave-One-Out Cross Validation (LOOCV), miRNA-fixed LOOCV, SM-fixed LOOCV and 5-fold cross-validation achieved excellent performance. Numerous hypothesized associations in case studies were confirmed by experimental literatures. All of these results confirmed that GCNLASMMA is a trustworthy association inference method.
Collapse
|
19
|
Peng L, Tu Y, Huang L, Li Y, Fu X, Chen X. DAESTB: inferring associations of small molecule-miRNA via a scalable tree boosting model based on deep autoencoder. Brief Bioinform 2022; 23:6827720. [PMID: 36377749 DOI: 10.1093/bib/bbac478] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) are closely related to a variety of human diseases, not only regulating gene expression, but also having an important role in human life activities and being viable targets of small molecule drugs for disease treatment. Current computational techniques to predict the potential associations between small molecule and miRNA are not that accurate. Here, we proposed a new computational method based on a deep autoencoder and a scalable tree boosting model (DAESTB), to predict associations between small molecule and miRNA. First, we constructed a high-dimensional feature matrix by integrating small molecule-small molecule similarity, miRNA-miRNA similarity and known small molecule-miRNA associations. Second, we reduced feature dimensionality on the integrated matrix using a deep autoencoder to obtain the potential feature representation of each small molecule-miRNA pair. Finally, a scalable tree boosting model is used to predict small molecule and miRNA potential associations. The experiments on two datasets demonstrated the superiority of DAESTB over various state-of-the-art methods. DAESTB achieved the best AUC value. Furthermore, in three case studies, a large number of predicted associations by DAESTB are confirmed with the public accessed literature. We envision that DAESTB could serve as a useful biological model for predicting potential small molecule-miRNA associations.
Collapse
Affiliation(s)
- Li Peng
- College of Computer Science and Engineering, Hunan University of Science and Technology, Xiangtan, 411201, Hunan, China.,Hunan Key Laboratory for Service computing and Novel Software Technology
| | - Yuan Tu
- College of Computer Science and Engineering, Hunan University of Science and Technology, Xiangtan, 411201, Hunan, China
| | - Li Huang
- Academy of Arts and Design, Tsinghua University, Beijing, 10084, China.,The Future Laboratory, Tsinghua University, Beijing, 10084, China
| | - Yang Li
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Xiangzheng Fu
- College of Information Science and Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Xiang Chen
- College of Computer Science and Engineering, Hunan University of Science and Technology, Xiangtan, 411201, Hunan, China
| |
Collapse
|
20
|
Dong TN, Schrader J, Mücke S, Khosla M. A message passing framework with multiple data integration for miRNA-disease association prediction. Sci Rep 2022; 12:16259. [PMID: 36171337 PMCID: PMC9519928 DOI: 10.1038/s41598-022-20529-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/14/2022] [Indexed: 11/08/2022] Open
Abstract
Micro RNA or miRNA is a highly conserved class of non-coding RNA that plays an important role in many diseases. Identifying miRNA-disease associations can pave the way for better clinical diagnosis and finding potential drug targets. We propose a biologically-motivated data-driven approach for the miRNA-disease association prediction, which overcomes the data scarcity problem by exploiting information from multiple data sources. The key idea is to enrich the existing miRNA/disease-protein-coding gene (PCG) associations via a message passing framework, followed by the use of disease ontology information for further feature filtering. The enriched and filtered PCG associations are then used to construct the inter-connected miRNA-PCG-disease network to train a structural deep network embedding (SDNE) model. Finally, the pre-trained embeddings and the biologically relevant features from the miRNA family and disease semantic similarity are concatenated to form the pair input representations to a Random Forest classifier whose task is to predict the miRNA-disease association probabilities. We present large-scale comparative experiments, ablation, and case studies to showcase our approach's superiority. Besides, we make the model prediction results for 1618 miRNAs and 3679 diseases, along with all related information, publicly available at http://software.mpm.leibniz-ai-lab.de/ to foster assessments and future adoption.
Collapse
Affiliation(s)
- Thi Ngan Dong
- L3S Research Center, Leibniz University of Hannover, Hannover, Germany.
| | - Johanna Schrader
- L3S Research Center, Leibniz University of Hannover, Hannover, Germany
| | - Stefanie Mücke
- Hannover Unified Biobank (HUB), Hannover Medical School, Hannover, Germany
| | - Megha Khosla
- Delft University of Technology (TU Delft), Delft, Netherlands
| |
Collapse
|
21
|
Huang L, Zhang L, Chen X. Updated review of advances in microRNAs and complex diseases: taxonomy, trends and challenges of computational models. Brief Bioinform 2022; 23:6686738. [PMID: 36056743 DOI: 10.1093/bib/bbac358] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 12/12/2022] Open
Abstract
Since the problem proposed in late 2000s, microRNA-disease association (MDA) predictions have been implemented based on the data fusion paradigm. Integrating diverse data sources gains a more comprehensive research perspective, and brings a challenge to algorithm design for generating accurate, concise and consistent representations of the fused data. After more than a decade of research progress, a relatively simple algorithm like the score function or a single computation layer may no longer be sufficient for further improving predictive performance. Advanced model design has become more frequent in recent years, particularly in the form of reasonably combing multiple algorithms, a process known as model fusion. In the current review, we present 29 state-of-the-art models and introduce the taxonomy of computational models for MDA prediction based on model fusion and non-fusion. The new taxonomy exhibits notable changes in the algorithmic architecture of models, compared with that of earlier ones in the 2017 review by Chen et al. Moreover, we discuss the progresses that have been made towards overcoming the obstacles to effective MDA prediction since 2017 and elaborated on how future models can be designed according to a set of new schemas. Lastly, we analysed the strengths and weaknesses of each model category in the proposed taxonomy and proposed future research directions from diverse perspectives for enhancing model performance.
Collapse
Affiliation(s)
- Li Huang
- Academy of Arts and Design, Tsinghua University, Beijing, 10084, China.,The Future Laboratory, Tsinghua University, Beijing, 10084, China
| | - Li Zhang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, China
| | - Xing Chen
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, China.,Artificial Intelligence Research Institute, China University of Mining and Technology, Xuzhou, 221116, China
| |
Collapse
|
22
|
Xiong Z, Huang F, Wang Z, Liu S, Zhang W. A Multimodal Framework for Improving in Silico Drug Repositioning With the Prior Knowledge From Knowledge Graphs. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2623-2631. [PMID: 34375284 DOI: 10.1109/tcbb.2021.3103595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Drug repositioning/repurposing is a very important approach towards identifying novel treatments for diseases in drug discovery. Recently, large-scale biological datasets are increasingly available for pharmaceutical research and promote the development of drug repositioning, but efficiently utilizing these datasets remains challenging. In this paper, we develop a novel multimodal framework, termed GraphPK (Graph-based Prior Knowledge) for improving in silico drug repositioning via using the prior knowledge from a drug knowledge graph. First, we construct a knowledge graph by integrating relevant bio-entities (drugs, diseases, etc.) and associations/interactions among them, and apply the knowledge graph embedding technique to extract prior knowledge of drugs and diseases. Moreover, we make use of the known drug-disease association, and obtain known association-based features from an association bipartite graph through graph embedding, and also take into account biological domain features, i.e., drug chemical structures and disease semantic similarity. Finally, we design a multimodal neural network to combine three types of features from the knowledge graph, the known associations and the biological domain, and build the prediction model for predicting drug-disease associations. Massive experiments show that our method outperforms other state-of-the-art methods in terms of most metrics, and the ablation analysis regarding the three types of features reveals that prior knowledge from knowledge graphs can not only lift the predictive power of in silico drug repositioning, but also enhance the model's robustness to different scenarios. The results of case studies offer support that GraphPK has the potential for actual use.
Collapse
|
23
|
Yu F, Li B, Sun J, Qi J, De Wilde RL, Torres-de la Roche LA, Li C, Ahmad S, Shi W, Li X, Chen Z. PSRR: A Web Server for Predicting the Regulation of miRNAs Expression by Small Molecules. Front Mol Biosci 2022; 9:817294. [PMID: 35386297 PMCID: PMC8979021 DOI: 10.3389/fmolb.2022.817294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Background: MicroRNAs (miRNAs) play key roles in a variety of pathological processes by interacting with their specific target mRNAs for translation repression and may function as oncogenes (oncomiRs) or tumor suppressors (TSmiRs). Therefore, a web server that could predict the regulation relations between miRNAs and small molecules is expected to achieve implications for identifying potential therapeutic targets for anti-tumor drug development. Methods: Upon obtaining positive/known small molecule-miRNA regulation pairs from SM2miR, we generated a multitude of high-quality negative/unknown pairs by leveraging similarities between the small molecule structures. Using the pool of the positive and negative pairs, we created the Dataset1 and Dataset2 datasets specific to up-regulation and down-regulation pairs, respectively. Manifold machine learning algorithms were then employed to construct models of predicting up-regulation and down-regulation pairs on the training portion of pairs in Dataset1 and Dataset2, respectively. Prediction abilities of the resulting models were further examined by discovering potential small molecules to regulate oncogenic miRNAs identified from miRNA sequencing data of endometrial carcinoma samples. Results: The random forest algorithm outperformed four machine-learning algorithms by achieving the highest AUC values of 0.911 for the up-regulation model and 0.896 for the down-regulation model on the testing datasets. Moreover, the down-regulation and up-regulation models yielded the accuracy values of 0.91 and 0.90 on independent validation pairs, respectively. In a case study, our model showed highly-reliable results by confirming all top 10 predicted regulation pairs as experimentally validated pairs. Finally, our predicted binding affinities of oncogenic miRNAs and small molecules bore a close resemblance to the lowest binding energy profiles using molecular docking. Predictions of the final model are freely accessible through the PSRR web server at https://rnadrug.shinyapps.io/PSRR/. Conclusion: Our study provides a novel web server that could effectively predict the regulation of miRNAs expression by small molecules.
Collapse
Affiliation(s)
- Fanrong Yu
- Department of Obstetrics and Gynecology, Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated to Sixth People’s Hospital South Campus, Shanghai, China
| | - Bihui Li
- Department of Oncology, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jianfeng Sun
- Department of Bioinformatics, Wissenschaftzentrum Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jing Qi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße, Düsseldorf, Germany
| | - Rudy Leon De Wilde
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | | | - Cheng Li
- Department of Orthopaedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Wenjie Shi
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Xiqing Li
- Oncology Department, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Zihao Chen
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| |
Collapse
|
24
|
Muslu O, Hoyt CT, Lacerda M, Hofmann-Apitius M, Frohlich H. GuiltyTargets: Prioritization of Novel Therapeutic Targets With Network Representation Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:491-500. [PMID: 32750869 DOI: 10.1109/tcbb.2020.3003830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The majority of clinical trials fail due to low efficacy of investigated drugs, often resulting from a poor choice of target protein. Existing computational approaches aim to support target selection either via genetic evidence or by putting potential targets into the context of a disease specific network reconstruction. The purpose of this work was to investigate whether network representation learning techniques could be used to allow for a machine learning based prioritization of putative targets. We propose a novel target prioritization approach, GuiltyTargets, which relies on attributed network representation learning of a genome-wide protein-protein interaction network annotated with disease-specific differential gene expression and uses positive-unlabeled (PU) machine learning for candidate ranking. We evaluated our approach on 12 datasets from six diseases of different type (cancer, metabolic, neurodegenerative) within a 10 times repeated 5-fold stratified cross-validation and achieved AUROC values between 0.92 - 0.97, significantly outperforming previous approaches that relied on manually engineered topological features. Moreover, we showed that GuiltyTargets allows for target repositioning across related disease areas. An application of GuiltyTargets to Alzheimer's disease resulted in a number of highly ranked candidates that are currently discussed as targets in the literature. Interestingly, one (COMT) is also the target of an approved drug (Tolcapone) for Parkinson's disease, highlighting the potential for target repositioning with our method. The GuiltyTargets Python package is available on PyPI and all code used for analysis can be found under the MIT License at https://github.com/GuiltyTargets. Attributed network representation learning techniques provide an interesting approach to effectively leverage the existing knowledge about the molecular mechanisms in different diseases. In this work, the combination with positive-unlabeled learning for target prioritization demonstrated a clear superiority compared to classical feature engineering approaches. Our work highlights the potential of attributed network representation learning for target prioritization. Given the overarching relevance of networks in computational biology we believe that attributed network representation learning techniques could have a broader impact in the future.
Collapse
|
25
|
Sun C, Xuan P, Zhang T, Ye Y. Graph Convolutional Autoencoder and Generative Adversarial Network-Based Method for Predicting Drug-Target Interactions. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:455-464. [PMID: 32750854 DOI: 10.1109/tcbb.2020.2999084] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The computational prediction of novel drug-target interactions (DTIs) may effectively speed up the process of drug repositioning and reduce its costs. Most previous methods integrated multiple kinds of connections about drugs and targets by constructing shallow prediction models. These methods failed to deeply learn the low-dimension feature vectors for drugs and targets and ignored the distribution of these feature vectors. We proposed a graph convolutional autoencoder and generative adversarial network (GAN)-based method, GANDTI, to predict DTIs. We constructed a drug-target heterogeneous network to integrate various connections related to drugs and targets, i.e., the similarities and interactions between drugs or between targets and the interactions between drugs and targets. A graph convolutional autoencoder was established to learn the network embeddings of the drug and target nodes in a low-dimensional feature space, and the autoencoder deeply integrated different kinds of connections within the network. A GAN was introduced to regularize the feature vectors of nodes into a Gaussian distribution. Severe class imbalance exists between known and unknown DTIs. Thus, we constructed a classifier based on an ensemble learning model, LightGBM, to estimate the interaction propensities of drugs and targets. This classifier completely exploited all unknown DTIs and counteracted the negative effect of class imbalance. The experimental results indicated that GANDTI outperforms several state-of-the-art methods for DTI prediction. Additionally, case studies of five drugs demonstrated the ability of GANDTI to discover the potential targets for drugs.
Collapse
|
26
|
Xie G, Li J, Gu G, Sun Y, Lin Z, Zhu Y, Wang W. BGMSDDA: a bipartite graph diffusion algorithm with multiple similarity integration for drug-disease association prediction. Mol Omics 2021; 17:997-1011. [PMID: 34610633 DOI: 10.1039/d1mo00237f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Drug repositioning, a method that relies on the information from the original drug-disease association matrix, aims to identify new indications for existing drugs and is expected to greatly reduce the cost and time of drug development. However, most current drug repositioning methods make use of the original drug-disease association matrix directly without preconditioning. As relatively only a few associations between drugs and diseases have been determined from actual observations, the original drug-disease association matrix used in the prediction is sparse, which affects the performance of the prediction method. A method for mining similar features of drugs and diseases is still lacking. To solve these problems, we developed a bipartite graph diffusion algorithm with multiple similarity integration for drug-disease association prediction (BGMSDDA). First, the weight K nearest known neighbors (WKNKN) algorithm was used to reconstruct the drug-disease association matrix. Secondly, an effective method was designed to extract similar characteristics of drugs and diseases based on integrating linear neighborhood similarity and Gaussian kernel similarity. Finally, bipartite graph diffusion was used to infer undiscovered drug-disease associations. After carrying out 10-fold cross-validation experiments, BGMSDDA showed excellent performance on two datasets, specifically with AUC values of 0.939 (Fdataset) and 0.954 (Cdataset), and AUPR values of 0.466 (Fdataset) and 0.565 (Cdataset). Furthermore, to evaluate the accuracy of the results of BGMSDDA, we conducted case studies on three medically used drugs selected from Fdataset and Cdataset and validated the predictive associated diseases of each drug with some databases. Based on the results obtained, BGMSDDA was demonstrated to be useful for predicting drug-disease associations.
Collapse
Affiliation(s)
- Guobo Xie
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| | - Jianming Li
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| | - Guosheng Gu
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| | - Yuping Sun
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| | - Zhiyi Lin
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| | - Yinting Zhu
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| | - Weiming Wang
- School of Computer Science, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
27
|
Wang W, Wang Y, Zhang Y, Liu D, Zhang H, Wang X. PPDTS: Predicting potential drug-target interactions based on network similarity. IET Syst Biol 2021; 16:18-27. [PMID: 34783172 PMCID: PMC8849239 DOI: 10.1049/syb2.12037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 11/19/2022] Open
Abstract
Identification of drug–target interactions (DTIs) has great practical importance in the drug discovery process for known diseases. However, only a small proportion of DTIs in these databases has been verified experimentally, and the computational methods for predicting the interactions remain challenging. As a result, some effective computational models have become increasingly popular for predicting DTIs. In this work, the authors predict potential DTIs from the local structure of drug–target associations' network, which is different from the traditional global network similarity methods based on structure and ligand. A novel method called PPDTS is proposed to predict DTIs. First, according to the DTIs’ network local structure, the known DTIs are converted into a binary network. Second, the Resource Allocation algorithm is used to obtain a drug–drug similarity network and a target–target similarity network. Third, a Collaborative Filtering algorithm is used with the known drug–target topology information to obtain similarity scores. Fourth, the linear combination of drug–target similarity model and the target–drug similarity model are innovatively proposed to obtain the final prediction results. Finally, the experimental performance of PPDTS has proved to be higher than that of the previously mentioned four popular network‐based similarity methods, which is validated in different experimental datasets. Some of the predicted results can be supported in UniProt and DrugBank databases.
Collapse
Affiliation(s)
- Wei Wang
- College of Computer and Information Engineering, Henan Normal University, Xinxiang, China.,Key Laboratory of Artificial Intelligence and Personalized Learning in Education of Henan Province, Henan Normal University, Xinxiang, China.,Big Data Engineering Laboratory for Teaching Resources and Assessment of Education Quality of Henan Province, Henan Normal University, Xinxiang, China
| | - Yongqing Wang
- College of Computer and Information Engineering, Henan Normal University, Xinxiang, China
| | - Yu Zhang
- College of Computer and Information Engineering, Henan Normal University, Xinxiang, China
| | - Dong Liu
- College of Computer and Information Engineering, Henan Normal University, Xinxiang, China.,Key Laboratory of Artificial Intelligence and Personalized Learning in Education of Henan Province, Henan Normal University, Xinxiang, China.,Big Data Engineering Laboratory for Teaching Resources and Assessment of Education Quality of Henan Province, Henan Normal University, Xinxiang, China
| | - Hongjun Zhang
- Computer Science and Technology, Anyang University, Anyang, China
| | - Xianfang Wang
- Computer Science and Technology, Henan Institute of Technology, Xinxiang, China
| |
Collapse
|
28
|
Luo J, Shen C, Lai Z, Cai J, Ding P. Incorporating Clinical, Chemical and Biological Information for Predicting Small Molecule-microRNA Associations Based on Non-Negative Matrix Factorization. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:2535-2545. [PMID: 32092012 DOI: 10.1109/tcbb.2020.2975780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Small molecule(SM) drugs can affect the expression of miRNAs, which plays crucial roles in many important biological processes. The chemical structure and clinical information of small molecule can simultaneously incorporate information such as anatomical distribution, therapeutic effects and structural characteristics. It is necessary to develop a novel model that incorporates small molecule chemical structure and clinical information to reveal the unknown small molecule-miRNA associations. In this study, we developed a new framework based on non-negative matrix factorization, called SMANMF, to discover the potential small molecules-miRNAs associations. First, the functional similarity of two miRNAs can be obtained by computing the overlap of the target gene sets in which the miRNAs interact together, and we integrated two types of small molecule similarities, including chemical similarity and clinical similarity. Then, we utilized a non-negative matrix factorization model to discover the unknown relationship between small molecules and miRNAs. The evaluation results indicate that our model can achieve superior prediction performance compared with previous approaches in 5-fold cross-validation. At the same time, the results of case studies also reveal that the SMANMF model has good predictive performance for predicting the potential association between small molecules and miRNAs.
Collapse
|
29
|
Xuan P, Chen B, Zhang T, Yang Y. Prediction of Drug-Target Interactions Based on Network Representation Learning and Ensemble Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:2671-2681. [PMID: 32340959 DOI: 10.1109/tcbb.2020.2989765] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Identifying interactions between drugs and target proteins is a critical step in the drug development process, as it helps identify new targets for drugs and accelerate drug development. The number of known drug-protein interactions (positive samples) is much lower than that of the unknown ones (negative samples), which forms a class imbalance. Most previous methods only utilised part of the negative samples to train the prediction model, so most of the information on negative samples was neglected. Therefore, a new method must be developed to predict candidate drug-related proteins and fully utilise negative samples to improve prediction performance. We present a method based on non-negative matrix factorisation and gradient boosting decision tree (GBDT), named NGDTP, to identify the candidate drug-protein interactions. NGDTP integrates multiple kinds of protein similarities, drugs-proteins interactions, and multiple kinds of drugs similarities at different levels, including target proteins of drugs, drug-related diseases, and side effects of drugs. We propose a network representation learning method based on matrix factorisation to learn low-dimensional vector representations of drug and protein nodes. On the basis of these low-dimensional node representations, a GBDT-based prediction model was constructed and it obtains the association scores through establishing multiple decision trees for a drug-protein pairs. NGDTP is an ensemble learning model that fully utilises all the negative samples to effectively alleviate the problem of class imbalance. NGDTP achieves superior prediction performance when it is compared with several state-of-the-art methods. The experimental results indicate that NGDTP also retrieves more actual drug-protein interactions in the top part of prediction result, which drew significant attention from the biologists. In addition, case studies on 10 drugs further confirmed the ability of the NGDTP to identify potential candidate proteins for drugs.
Collapse
|
30
|
Ding P, Ouyang W, Luo J, Kwoh CK. Heterogeneous information network and its application to human health and disease. Brief Bioinform 2021; 21:1327-1346. [PMID: 31566212 DOI: 10.1093/bib/bbz091] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 12/11/2022] Open
Abstract
The molecular components with the functional interdependencies in human cell form complicated biological network. Diseases are mostly caused by the perturbations of the composite of the interaction multi-biomolecules, rather than an abnormality of a single biomolecule. Furthermore, new biological functions and processes could be revealed by discovering novel biological entity relationships. Hence, more and more biologists focus on studying the complex biological system instead of the individual biological components. The emergence of heterogeneous information network (HIN) offers a promising way to systematically explore complicated and heterogeneous relationships between various molecules for apparently distinct phenotypes. In this review, we first present the basic definition of HIN and the biological system considered as a complex HIN. Then, we discuss the topological properties of HIN and how these can be applied to detect network motif and functional module. Afterwards, methodologies of discovering relationships between disease and biomolecule are presented. Useful insights on how HIN aids in drug development and explores human interactome are provided. Finally, we analyze the challenges and opportunities for uncovering combinatorial patterns among pharmacogenomics and cell-type detection based on single-cell genomic data.
Collapse
Affiliation(s)
- Pingjian Ding
- School of Computer Science, University of South China, Hengyang, China
| | - Wenjue Ouyang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Chee-Keong Kwoh
- School of Computer Science and Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
31
|
Chen X, Zhou C, Wang CC, Zhao Y. Predicting potential small molecule-miRNA associations based on bounded nuclear norm regularization. Brief Bioinform 2021; 22:6353837. [PMID: 34404088 DOI: 10.1093/bib/bbab328] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Mounting evidence has demonstrated the significance of taking microRNAs (miRNAs) as the target of small molecule (SM) drugs for disease treatment. Given the fact that exploring new SM-miRNA associations through biological experiments is extremely expensive, several computing models have been constructed to reveal the possible SM-miRNA associations. Here, we built a computing model of Bounded Nuclear Norm Regularization for SM-miRNA Associations prediction (BNNRSMMA). Specifically, we first constructed a heterogeneous SM-miRNA network utilizing miRNA similarity, SM similarity, confirmed SM-miRNA associations and defined a matrix to represent the heterogeneous network. Then, we constructed a model to complete this matrix by minimizing its nuclear norm. The Alternating Direction Method of Multipliers was adopted to minimize the nuclear norm and obtain predicted scores. The main innovation lies in two aspects. During completion, we limited all elements of the matrix within the interval of (0,1) to make sure they have practical significance. Besides, instead of strictly fitting all known elements, a regularization term was incorporated to tolerate the noise in integrated similarities. Furthermore, four kinds of cross-validations on two datasets and two types of case studies were performed to evaluate the predictive performance of BNNRSMMA. Finally, BNNRSMMA attained areas under the curve of 0.9822 (0.8433), 0.9793 (0.8852), 0.8253 (0.7350) and 0.9758 ± 0.0029 (0.8759 ± 0.0041) under global leave-one-out cross-validation (LOOCV), miRNA-fixed LOOCV, SM-fixed LOOCV and 5-fold cross-validation based on Dataset 1(Dataset 2), respectively. With regard to case studies, plenty of predicted associations have been verified by experimental literatures. All these results confirmed that BNNRSMMA is a reliable tool for inferring associations.
Collapse
Affiliation(s)
- Xing Chen
- Artificial Intelligence Research Institute, China University of Mining and Technology, Xuzhou 221116, China
| | - Chi Zhou
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Chun-Chun Wang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Yan Zhao
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| |
Collapse
|
32
|
Ding P, Liang C, Ouyang W, Li G, Xiao Q, Luo J. Inferring Synergistic Drug Combinations Based on Symmetric Meta-Path in a Novel Heterogeneous Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1562-1571. [PMID: 31714232 DOI: 10.1109/tcbb.2019.2951557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Combinatorial drug therapy is a promising way for treating cancers, which can reduce drug side effects and improve drug efficacy. However, due to the large-scale combinatorial space, it is difficult to quickly and effectively identify novel synergistic drug combinations for further implementing combinatorial drug therapy. The computational method of fusing multi-source knowledge is a time- and cost-efficient strategy to infer synergistic drug combinations for testing. However, for the existing computational methods of inferring synergistic drug combinations, it still remains a challenging to effectively combine multi-source information to achieve the desired results. Hence, in this study, we developed a novel Inference method of Synergistic Drug Combinations based on Symmetric Meta-Path (ISDCSMP), which can systematically and accurately prioritize synergistic drug combinations in a novel drug-target heterogeneous network integrating multi-source information. In the experiment, ISDCSMP outperformed the state-of-the-art methods in terms of AUC and precision on the benchmark dataset in five-fold cross validation. Moreover, we further illustrated performances of different ways for obtaining the combination coefficients, and analyzed the influences of the maximum meta-path length. The performances of various single meta-paths were described in five-fold cross validation. Finally, we confirmed the practical usefulness of ISDCSMP with the predicted novel synergistic drug combinations. The source code of ISDCSMP is available at https://github.com/KDDing/ISDCSMP.
Collapse
|
33
|
Pliakos K, Vens C, Tsoumakas G. Predicting Drug-Target Interactions With Multi-Label Classification and Label Partitioning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1596-1607. [PMID: 31689203 DOI: 10.1109/tcbb.2019.2951378] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Identifying drug-target interactions is crucial for drug discovery. Despite modern technologies used in drug screening, experimental identification of drug-target interactions is an extremely demanding task. Predicting drug-target interactions in silico can thereby facilitate drug discovery as well as drug repositioning. Various machine learning models have been developed over the years to predict such interactions. Multi-output learning models in particular have drawn the attention of the scientific community due to their high predictive performance and computational efficiency. These models are based on the assumption that all the labels are correlated with each other. However, this assumption is too optimistic. Here, we address drug-target interaction prediction as a multi-label classification task that is combined with label partitioning. We show that building multi-output learning models over groups (clusters) of labels often leads to superior results. The performed experiments confirm the efficiency of the proposed framework.
Collapse
|
34
|
Kaushik AC, Mehmood A, Selvaraj G, Dai X, Pan Y, Wei DQ. CoronaPep: An Anti-Coronavirus Peptide Generation Tool. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1299-1304. [PMID: 33687847 PMCID: PMC8769015 DOI: 10.1109/tcbb.2021.3064630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/02/2020] [Accepted: 09/14/2020] [Indexed: 02/05/2023]
Abstract
The novel coronavirus (COVID-19) infections have adopted the shape of a global pandemic now, demanding an urgent vaccine design. The current work reports contriving an anti-coronavirus peptide scanner tool to discern anti-coronavirus targets in the embodiment of peptides. The proffered CoronaPep tool features the fast fingerprinting of the anti-coronavirus target serving supreme prominence in the current bioinformatics research. The anti-coronavirus target protein sequences reported from the current outbreak are scanned against the anti-coronavirus target data-sets via CORONAPEP which provides precision-based anti-coronavirus peptides. This tool is specifically for the coronavirus data, which can predict peptides from the whole genome, or a gene or protein's list. Besides it is relatively fast, accurate, userfriendly and can generate maximum output from the limited information. The availability of tools like CORONAPEP will immeasurably perquisite researchers in the discipline of oncology and structure-based drug design.
Collapse
Affiliation(s)
| | - Aamir Mehmood
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
- Peng Cheng LaboratoryShenzhenGuangdong518055China
| | - Gurudeeban Selvaraj
- Center of Interdisciplinary Sciences-Computational Life Sciences, College of Food Science and EngineeringHenan University of TechnologyZhengzhou450001China
- Centre for Research in Molecular ModelingConcordia UniversityMontrealQCH4B 1R6Canada
| | - Xiaofeng Dai
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
| | - Yi Pan
- Department of Computer ScienceGeorgia State UniversityAtlantaGA30302-5060USA
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
- Peng Cheng LaboratoryShenzhenGuangdong518055China
| |
Collapse
|
35
|
Li J, Peng D, Xie Y, Dai Z, Zou X, Li Z. Novel Potential Small Molecule-MiRNA-Cancer Associations Prediction Model Based on Fingerprint, Sequence, and Clinical Symptoms. J Chem Inf Model 2021; 61:2208-2219. [PMID: 33899462 DOI: 10.1021/acs.jcim.0c01458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
As an important biomarker in organisms, miRNA is closely related to various small molecules and diseases. Research on small molecule-miRNA-cancer associations is helpful for the development of cancer treatment drugs and the discovery of pathogenesis. It is very urgent to develop theoretical methods for identifying potential small molecular-miRNA-cancer associations, because experimental approaches are usually time-consuming, laborious, and expensive. To overcome this problem, we developed a new computational method, in which features derived from structure, sequence, and symptoms were utilized to characterize small molecule, miRNA, and cancer, respectively. A feature vector was construct to characterize small molecule-miRNA-cancer association by concatenating these features, and a random forest algorithm was utilized to construct a model for recognizing potential association. Based on the 5-fold cross-validation and benchmark data set, the model achieved an accuracy of 93.20 ± 0.52%, a precision of 93.22 ± 0.51%, a recall of 93.20 ± 0.53%, and an F1-measure of 93.20 ± 0.52%. The areas under the receiver operating characteristic curve and precision recall curve were 0.9873 and 0.9870. The real prediction ability and application performance of the developed method have also been further evaluated and verified through an independent data set test and case study. Some potential small molecules and miRNAs related to cancer have been identified and are worthy of further experimental research. It is anticipated that our model could be regarded as a useful high-throughput virtual screening tool for drug research and development. All source codes can be downloaded from https://github.com/LeeKamlong/Multi-class-SMMCA.
Collapse
Affiliation(s)
- Jinlong Li
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
| | - Dongdong Peng
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
| | - Yun Xie
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
| | - Zong Dai
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Xiaoyong Zou
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Zhanchao Li
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of Traditional Chinese Medicine, Guangzhou 510006, People's Republic of China
| |
Collapse
|
36
|
Shen C, Luo J, Ouyang W, Ding P, Wu H. Identification of Small Molecule–miRNA Associations with Graph Regularization Techniques in Heterogeneous Networks. J Chem Inf Model 2020; 60:6709-6721. [DOI: 10.1021/acs.jcim.0c00975] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Cong Shen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Wenjue Ouyang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Pingjian Ding
- School of Computer Science, University of South China, Hengyang 421001, China
| | - Hao Wu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| |
Collapse
|
37
|
Xu P, Wu Q, Lu D, Yu J, Rao Y, Kou Z, Fang G, Liu W, Han H. A systematic study of critical miRNAs on cells proliferation and apoptosis by the shortest path. BMC Bioinformatics 2020; 21:396. [PMID: 32894041 PMCID: PMC7487489 DOI: 10.1186/s12859-020-03732-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MicroRNAs are a class of important small noncoding RNAs, which have been reported to be involved in the processes of tumorigenesis and development by targeting a few genes. Existing studies show that the imbalance between cell proliferation and apoptosis is closely related to the initiation and development of cancers. However, the impact of miRNAs on this imbalance has not been studied systematically. RESULTS In this study, we first construct a cell fate miRNA-gene regulatory network. Then, we propose a systematical method for calculating the global impact of miRNAs on cell fate genes based on the shortest path. Results on breast cancer and liver cancer datasets show that most of the cell fate genes are perturbed by the differentially expressed miRNAs. Most of the top-identified miRNAs are verified in the Human MicroRNA Disease Database (HMDD) and are related to breast and liver cancers. Function analysis shows that the top 20 miRNAs regulate multiple cell fate related function modules and interact tightly based on their functional similarity. Furthermore, more than half of them can promote sensitivity or induce resistance to some anti-cancer drugs. Besides, survival analysis demonstrates that the top-ranked miRNAs are significantly related to the overall survival time in the breast and liver cancers group. CONCLUSION In sum, this study can help to systematically study the important role of miRNAs on proliferation and apoptosis and thereby uncover the key miRNAs during the process of tumorigenesis. Furthermore, the results of this study will contribute to the development of clinical therapy based miRNAs for cancers.
Collapse
Affiliation(s)
- Peng Xu
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China.,School of computer science of information technology, Qiannan Normal University for Nationalities, Duyun, 558000, Guizhou, China
| | - Qian Wu
- College of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou, 325035, China
| | - Deyang Lu
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Jian Yu
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Yongsheng Rao
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Zheng Kou
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Gang Fang
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Wenbin Liu
- Institute of computational science and technology, Guangzhou University, Guangzhou, 510006, Guangdong, China.
| | - Henry Han
- Department of Computer and Information Science, Fordham University, New York, NY, 10023, USA.
| |
Collapse
|
38
|
Wong L, You ZH, Guo ZH, Yi HC, Chen ZH, Cao MY. MIPDH: A Novel Computational Model for Predicting microRNA-mRNA Interactions by DeepWalk on a Heterogeneous Network. ACS OMEGA 2020; 5:17022-17032. [PMID: 32715187 PMCID: PMC7376568 DOI: 10.1021/acsomega.9b04195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 03/06/2020] [Indexed: 06/11/2023]
Abstract
Analysis of miRNA-target mRNA interaction (MTI) is of crucial significance in discovering new target candidates for miRNAs. However, the biological experiments for identifying MTIs have a high false positive rate and are high-priced, time-consuming, and arduous. It is an urgent task to develop effective computational approaches to enhance the investigation of miRNA-target mRNA relationships. In this study, a novel method called MIPDH is developed for miRNA-mRNA interaction prediction by using DeepWalk on a heterogeneous network. More specifically, MIPDH extracts two kinds of features, in which a biological behavior feature is learned using a network embedding algorithm on a constructed heterogeneous network derived from 17 kinds of associations among drug, disease, and 6 kinds of biomolecules, and the attribute feature is learned using the k-mer method on sequences of miRNAs and target mRNAs. Then, a random forest classifier is trained on the features combined with the biological behavior feature and attribute feature. When implementing a 5-fold cross-validation experiment, MIPDH achieved an average accuracy, sensitivity, specificity and AUC of 75.85, 74.37, 77.33%, and 0.8044, respectively. To further evaluate the performance of MIPDH, other classifiers and feature descriptors are conducted for comparisons. MIPDH can achieve a better performance. Additionally, case studies on hsa-miR-106b-5p, hsa-let-7d-5p, and hsa-let-7e-5p are also implemented. As a result, 14, 9, and 9 out of the top 15 targets that interacted with these miRNAs were verified using the experimental literature or other databases. All these prediction results indicate that MIPDH is an effective method for predicting miRNA-target mRNA interactions.
Collapse
Affiliation(s)
- Leon Wong
- The
Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- XinJiang
Laboratory of Minority Speech and Language Information Processing, Chinese Academy of Sciences, Urumqi 830011, China
| | - Zhu-Hong You
- The
Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- XinJiang
Laboratory of Minority Speech and Language Information Processing, Chinese Academy of Sciences, Urumqi 830011, China
| | - Zhen-Hao Guo
- The
Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- XinJiang
Laboratory of Minority Speech and Language Information Processing, Chinese Academy of Sciences, Urumqi 830011, China
| | - Hai-Cheng Yi
- The
Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- XinJiang
Laboratory of Minority Speech and Language Information Processing, Chinese Academy of Sciences, Urumqi 830011, China
| | - Zhan-Heng Chen
- The
Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- XinJiang
Laboratory of Minority Speech and Language Information Processing, Chinese Academy of Sciences, Urumqi 830011, China
| | - Mei-Yuan Cao
- Guang
Dong Polytechnic College, Zhaoqing 526100, Guangdong, China
| |
Collapse
|
39
|
Shen C, Luo J, Lai Z, Ding P. Multiview Joint Learning-Based Method for Identifying Small-Molecule-Associated MiRNAs by Integrating Pharmacological, Genomics, and Network Knowledge. J Chem Inf Model 2020; 60:4085-4097. [DOI: 10.1021/acs.jcim.0c00244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Cong Shen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Zihan Lai
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Pingjian Ding
- School of Computer Science, University of South China, Hengyang 421001, China
| |
Collapse
|
40
|
Drug repositioning based on the target microRNAs using bilateral-inductive matrix completion. Mol Genet Genomics 2020; 295:1305-1314. [DOI: 10.1007/s00438-020-01702-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
|
41
|
Wang CC, Zhao Y, Chen X. Drug-pathway association prediction: from experimental results to computational models. Brief Bioinform 2020; 22:5835554. [PMID: 32393976 DOI: 10.1093/bib/bbaa061] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Effective drugs are urgently needed to overcome human complex diseases. However, the research and development of novel drug would take long time and cost much money. Traditional drug discovery follows the rule of one drug-one target, while some studies have demonstrated that drugs generally perform their task by affecting related pathway rather than targeting single target. Thus, the new strategy of drug discovery, namely pathway-based drug discovery, have been proposed. Obviously, identifying associations between drugs and pathways plays a key role in the development of pathway-based drug discovery. Revealing the drug-pathway associations by experiment methods would take much time and cost. Therefore, some computational models were established to predict potential drug-pathway associations. In this review, we first introduced the background of drug and the concept of drug-pathway associations. Then, some publicly accessible databases and web servers about drug-pathway associations were listed. Next, we summarized some state-of-the-art computational methods in the past years for inferring drug-pathway associations and divided these methods into three classes, namely Bayesian spare factor-based, matrix decomposition-based and other machine learning methods. In addition, we introduced several evaluation strategies to estimate the predictive performance of various computational models. In the end, we discussed the advantages and limitations of existing computational methods and provided some suggestions about the future directions of the data collection and the calculation models development.
Collapse
|
42
|
Liu F, Peng L, Tian G, Yang J, Chen H, Hu Q, Liu X, Zhou L. Identifying Small Molecule-miRNA Associations Based on Credible Negative Sample Selection and Random Walk. Front Bioeng Biotechnol 2020; 8:131. [PMID: 32258003 PMCID: PMC7090022 DOI: 10.3389/fbioe.2020.00131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/10/2020] [Indexed: 12/05/2022] Open
Abstract
Recently, many studies have demonstrated that microRNAs (miRNAs) are new small molecule drug targets. Identifying small molecule-miRNA associations (SMiRs) plays an important role in finding new clues for various human disease therapy. Wet experiments can discover credible SMiR associations; however, this is a costly and time-consuming process. Computational models have therefore been developed to uncover possible SMiR associations. In this study, we designed a new SMiR association prediction model, RWNS. RWNS integrates various biological information, credible negative sample selections, and random walk on a triple-layer heterogeneous network into a unified framework. It includes three procedures: similarity computation, negative sample selection, and SMiR association prediction based on random walk on the constructed small molecule-disease-miRNA association network. To evaluate the performance of RWNS, we used leave-one-out cross-validation (LOOCV) and 5-fold cross validation to compare RWNS with two state-of-the-art SMiR association methods, namely, TLHNSMMA and SMiR-NBI. Experimental results showed that RWNS obtained an AUC value of 0.9829 under LOOCV and 0.9916 under 5-fold cross validation on the SM2miR1 dataset, and it obtained an AUC value of 0.8938 under LOOCV and 0.9899 under 5-fold cross validation on the SM2miR2 dataset. More importantly, RWNS successfully captured 9, 17, and 37 SMiR associations validated by experiments among the predicted top 10, 20, and 50 SMiR candidates with the highest scores, respectively. We inferred that enoxacin and decitabine are associated with mir-21 and mir-155, respectively. Therefore, RWNS can be a powerful tool for SMiR association prediction.
Collapse
Affiliation(s)
- Fuxing Liu
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Geng Tian
- Geneis (Beijing) Co. Ltd., Beijing, China
| | | | - Hui Chen
- College of Chemical Engineering, Xiangtan University, Xiangtan, China
| | - Qi Hu
- Xiangya Second Hospital, Central South University, Changsha, Hunan, China
| | - Xiaojun Liu
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| |
Collapse
|
43
|
Ahmed F, Ijaz B, Ahmad Z, Farooq N, Sarwar MB, Husnain T. Modification of miRNA Expression through plant extracts and compounds against breast cancer: Mechanism and translational significance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 68:153168. [PMID: 31982837 DOI: 10.1016/j.phymed.2020.153168] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/02/2020] [Accepted: 01/04/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Cancer is hyper-proliferative, multi-factorial and multi-step, heterogeneous group of molecular disorders. It is the second most reported disease after heart diseases. Breast carcinoma is the foremost death causing disease in female population worldwide. Cancer can be controlled by regulating the gene expression. Current therapeutic options are associated with severe side effects and are expensive for the people living in under-developed countries. Plant derived substances have potential application against different diseases like cancer, inflammation and viral infections. HYPOTHESIS The mechanism of action of the medicinal plants is largely unknown. Targeting gene network and miRNA using medicinal plants could help in improving the therapeutic options against cancer. METHODS The literature from 135 articles was reviewed by using PubMed, google scholar, Science direct to find out the plants and plant-based compounds against breast cancer and also the studies reporting their mechanistic route of action both at coding and noncoding RNA levels. RESULTS Natural products act as selective inhibitors of the cancerous cells by targeting oncogenes and tumor suppressor genes or altering miRNA expression. Natural compounds like EGCG from tea, Genistein from fava beans, curcumin from turmeric, DIM found in cruciferous, Resveratrol a polyphenol and Quercetin a flavonoid is found in various plants have been studied for their anticancer activity. The EGCG was found to inhibit proliferative activity by modulating miR-16 and miR-21. Similarly, DIM was found to down regulate miR-92a which results to modulate NFkB and stops cancer development. Another plant-based compound Glyceollins found to upregulate miR-181c and miR-181d having role in tumor suppression. It also found to regulate miR-22, 29b and c, miR-30d, 34a and 195. Quercetin having anti-cancer activity induce the apoptosis through regulating miR-16, 26b, 34a, let-7g, 125a and miR-605 and reduce the miRNA expression like miR-146a/b, 503 and 194 which are involved in metastasis. CONCLUSION Targeting miRNA expression using natural plant extracts can have a reverse effect on cell proliferation; turning on and off tumor-inducing and suppressing genes. It can be efficiently adopted as an adjuvant with the conventional form of therapies to increase their efficacy against cancer progression.
Collapse
Affiliation(s)
- Fayyaz Ahmed
- National Center of Excellence in Molecular Biology, University of the Punjab Lahore, Pakistan
| | - Bushra Ijaz
- National Center of Excellence in Molecular Biology, University of the Punjab Lahore, Pakistan.
| | - Zarnab Ahmad
- National Center of Excellence in Molecular Biology, University of the Punjab Lahore, Pakistan
| | - Nadia Farooq
- Department of Surgery, Sir Gangaram Hospital Lahore Punjab, Pakistan
| | - Muhammad Bilal Sarwar
- National Center of Excellence in Molecular Biology, University of the Punjab Lahore, Pakistan
| | - Tayyab Husnain
- National Center of Excellence in Molecular Biology, University of the Punjab Lahore, Pakistan
| |
Collapse
|
44
|
Associating lncRNAs with small molecules via bilevel optimization reveals cancer-related lncRNAs. PLoS Comput Biol 2019; 15:e1007540. [PMID: 31877126 PMCID: PMC6948815 DOI: 10.1371/journal.pcbi.1007540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/08/2020] [Accepted: 11/12/2019] [Indexed: 12/28/2022] Open
Abstract
Long noncoding RNA (lncRNA) transcripts have emerging impacts in cancer studies, which suggests their potential as novel therapeutic agents. However, the molecular mechanism behind their treatment effects is still unclear. Here, we designed a computational model to Associate LncRNAs with Anti-Cancer Drugs (ALACD) based on a bilevel optimization model, which optimized the gene signature overlap in the upper level and imputed the missing lncRNA-gene association in the lower level. ALACD predicts genes coexpressed with lncRNAs mean while matching drug’s gene signatures. This model allows us to borrow the target gene information of small molecules to understand the mechanisms of action of lncRNAs and their roles in cancer. The ALACD model was systematically applied to the 10 cancer types in The Cancer Genome Atlas (TCGA) that had matched lncRNA and mRNA expression data. Cancer type-specific lncRNAs and associated drugs were identified. These lncRNAs show significantly different expression levels in cancer patients. Follow-up functional and molecular pathway analysis suggest the gene signatures bridging drugs and lncRNAs are closely related to cancer development. Importantly, patient survival information and evidence from the literature suggest that the lncRNAs and drug-lncRNA associations identified by the ALACD model can provide an alternative choice for cancer targeting treatment and potential cancer pognostic biomarkers. The ALACD model is freely available at https://github.com/wangyc82/ALACD-v1. LncRNAs are RNA transcripts that are longer than 200 bp and do not encode proteins. Recent experimental studies have indicated the crucial role of lncRNAs in cancer. We proposed a computational model, ALACD, to understand a lncRNA’s molecular mechanism by associating it with a drug through the drug’s target genes. ALACD reveals lncRNAs, the associated anti-cancer drug, and the induced gene signatures that are involved in the regulation of cancer. Furthermore, these cancer-related lncRNAs are differentially expressed in cancer patients and closely associated with patient survival.
Collapse
|
45
|
Chu Y, Kaushik AC, Wang X, Wang W, Zhang Y, Shan X, Salahub DR, Xiong Y, Wei DQ. DTI-CDF: a cascade deep forest model towards the prediction of drug-target interactions based on hybrid features. Brief Bioinform 2019; 22:451-462. [PMID: 31885041 DOI: 10.1093/bib/bbz152] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022] Open
Abstract
Drug-target interactions (DTIs) play a crucial role in target-based drug discovery and development. Computational prediction of DTIs can effectively complement experimental wet-lab techniques for the identification of DTIs, which are typically time- and resource-consuming. However, the performances of the current DTI prediction approaches suffer from a problem of low precision and high false-positive rate. In this study, we aim to develop a novel DTI prediction method for improving the prediction performance based on a cascade deep forest (CDF) model, named DTI-CDF, with multiple similarity-based features between drugs and the similarity-based features between target proteins extracted from the heterogeneous graph, which contains known DTIs. In the experiments, we built five replicates of 10-fold cross-validation under three different experimental settings of data sets, namely, corresponding DTI values of certain drugs (SD), targets (ST), or drug-target pairs (SP) in the training sets are missed but existed in the test sets. The experimental results demonstrate that our proposed approach DTI-CDF achieves a significantly higher performance than that of the traditional ensemble learning-based methods such as random forest and XGBoost, deep neural network, and the state-of-the-art methods such as DDR. Furthermore, there are 1352 newly predicted DTIs which are proved to be correct by KEGG and DrugBank databases. The data sets and source code are freely available at https://github.com//a96123155/DTI-CDF.
Collapse
Affiliation(s)
- Yanyi Chu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University
| | | | - Xiangeng Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University
| | - Wei Wang
- Mathematical Sciences, Shanghai Jiao Tong University
| | - Yufang Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University
| | | | | | - Yi Xiong
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University
| | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University
| |
Collapse
|
46
|
Yang M, Luo H, Li Y, Wu FX, Wang J. Overlap matrix completion for predicting drug-associated indications. PLoS Comput Biol 2019; 15:e1007541. [PMID: 31869322 PMCID: PMC6946175 DOI: 10.1371/journal.pcbi.1007541] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 01/07/2020] [Accepted: 11/12/2019] [Indexed: 11/19/2022] Open
Abstract
Identification of potential drug–associated indications is critical for either approved or novel drugs in drug repositioning. Current computational methods based on drug similarity and disease similarity have been developed to predict drug–disease associations. When more reliable drug- or disease-related information becomes available and is integrated, the prediction precision can be continuously improved. However, it is a challenging problem to effectively incorporate multiple types of prior information, representing different characteristics of drugs and diseases, to identify promising drug–disease associations. In this study, we propose an overlap matrix completion (OMC) for bilayer networks (OMC2) and tri-layer networks (OMC3) to predict potential drug-associated indications, respectively. OMC is able to efficiently exploit the underlying low-rank structures of the drug–disease association matrices. In OMC2, first of all, we construct one bilayer network from drug-side aspect and one from disease-side aspect, and then obtain their corresponding block adjacency matrices. We then propose the OMC2 algorithm to fill out the values of the missing entries in these two adjacency matrices, and predict the scores of unknown drug–disease pairs. Moreover, we further extend OMC2 to OMC3 to handle tri-layer networks. Computational experiments on various datasets indicate that our OMC methods can effectively predict the potential drug–disease associations. Compared with the other state-of-the-art approaches, our methods yield higher prediction accuracy in 10-fold cross-validation and de novo experiments. In addition, case studies also confirm the effectiveness of our methods in identifying promising indications for existing drugs in practical applications. This work introduces a computational approach, namely overlap matrix completion (OMC), to predict potential associations between drugs and diseases. The novelty of OMC lies in constructing an efficient framework of incorporating multiple types of prior information in bilayer and tri-layer networks. OMC for bilayer networks (OMC2) can approximate the low-rank structures of the drug–disease association matrices from both drug-side and disease-side. In addition, we further improve the prediction accuracy by extending OMC to handle tri-layer networks and develop its corresponding algorithm (OMC3). To evaluate the performance of OMC2 and OMC3, we conduct 10-fold cross-validation and de novo experiments on three datasets. Our computational results demonstrate that both OMC2 and OMC3 generally outperform five state-of-the-art methods in terms of ROC curve, PR curve, and top-ranked predictions.
Collapse
Affiliation(s)
- Mengyun Yang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R. China
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang, Hunan, P.R. China
| | - Huimin Luo
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R. China
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, Virginia, United States of America
| | - Fang-Xiang Wu
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R. China
- * E-mail:
| |
Collapse
|
47
|
Wang CC, Chen X. A Unified Framework for the Prediction of Small Molecule–MicroRNA Association Based on Cross-Layer Dependency Inference on Multilayered Networks. J Chem Inf Model 2019; 59:5281-5293. [DOI: 10.1021/acs.jcim.9b00667] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Chun-Chun Wang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Xing Chen
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| |
Collapse
|
48
|
To KKW, Fong W, Tong CWS, Wu M, Yan W, Cho WCS. Advances in the discovery of microRNA-based anticancer therapeutics: latest tools and developments. Expert Opin Drug Discov 2019; 15:63-83. [PMID: 31739699 DOI: 10.1080/17460441.2020.1690449] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: MicroRNAs (miRNAs) are small endogenous non-coding RNAs that repress the expression of their target genes by reducing mRNA stability and/or inhibiting translation. miRNAs are known to be aberrantly regulated in cancers. Modulators of miRNA (mimics and antagonists) have emerged as novel therapeutic tools for cancer treatment.Areas covered: This review summarizes the various strategies that have been applied to correct the dysregulated miRNA in cancer cells. The authors also discuss the recent advances in the technical development and preclinical/clinical evaluation of miRNA-based therapeutic agents.Expert opinion: Application of miRNA-based therapeutics for cancer treatment is appealing because they are able to modulate multiple dysregulated genes and/or signaling pathways in cancer cells. Major obstacles hindering their clinical development include drug delivery, off-target effects, efficacious dose determination, and safety. Tumor site-specific delivery of novel miRNA therapeutics may help to minimize off-target effects and toxicity. Combination of miRNA therapeutics with other anticancer treatment modalities could provide a synergistic effect, thus allowing the use of lower dose, minimizing off-target effects, and improving the overall safety profile in cancer patients. It is critical to identify individual miRNAs with cancer type-specific and context-specific regulation of oncogenes and tumor-suppressor genes in order to facilitate the precise use of miRNA anticancer therapeutics.
Collapse
Affiliation(s)
- Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Winnie Fong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Christy W S Tong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mingxia Wu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wei Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| |
Collapse
|
49
|
Network-based Biased Tree Ensembles (NetBiTE) for Drug Sensitivity Prediction and Drug Sensitivity Biomarker Identification in Cancer. Sci Rep 2019; 9:15918. [PMID: 31685861 PMCID: PMC6828742 DOI: 10.1038/s41598-019-52093-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/07/2019] [Indexed: 12/15/2022] Open
Abstract
We present the Network-based Biased Tree Ensembles (NetBiTE) method for drug sensitivity prediction and drug sensitivity biomarker identification in cancer using a combination of prior knowledge and gene expression data. Our devised method consists of a biased tree ensemble that is built according to a probabilistic bias weight distribution. The bias weight distribution is obtained from the assignment of high weights to the drug targets and propagating the assigned weights over a protein-protein interaction network such as STRING. The propagation of weights, defines neighborhoods of influence around the drug targets and as such simulates the spread of perturbations within the cell, following drug administration. Using a synthetic dataset, we showcase how application of biased tree ensembles (BiTE) results in significant accuracy gains at a much lower computational cost compared to the unbiased random forests (RF) algorithm. We then apply NetBiTE to the Genomics of Drug Sensitivity in Cancer (GDSC) dataset and demonstrate that NetBiTE outperforms RF in predicting IC50 drug sensitivity, only for drugs that target membrane receptor pathways (MRPs): RTK, EGFR and IGFR signaling pathways. We propose based on the NetBiTE results, that for drugs that inhibit MRPs, the expression of target genes prior to drug administration is a biomarker for IC50 drug sensitivity following drug administration. We further verify and reinforce this proposition through control studies on, PI3K/MTOR signaling pathway inhibitors, a drug category that does not target MRPs, and through assignment of dummy targets to MRP inhibiting drugs and investigating the variation in NetBiTE accuracy.
Collapse
|
50
|
Li C, Liu H, Hu Q, Que J, Yao J. A Novel Computational Model for Predicting microRNA-Disease Associations Based on Heterogeneous Graph Convolutional Networks. Cells 2019; 8:cells8090977. [PMID: 31455028 PMCID: PMC6769654 DOI: 10.3390/cells8090977] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 01/13/2023] Open
Abstract
Identifying the interactions between disease and microRNA (miRNA) can accelerate drugs development, individualized diagnosis, and treatment for various human diseases. However, experimental methods are time-consuming and costly. So computational approaches to predict latent miRNA-disease interactions are eliciting increased attention. But most previous studies have mainly focused on designing complicated similarity-based methods to predict latent interactions between miRNAs and diseases. In this study, we propose a novel computational model, termed heterogeneous graph convolutional network for miRNA-disease associations (HGCNMDA), which is based on known human protein-protein interaction (PPI) and integrates four biological networks: miRNA-disease, miRNA-gene, disease-gene, and PPI network. HGCNMDA achieved reliable performance using leave-one-out cross-validation (LOOCV). HGCNMDA is then compared to three state-of-the-art algorithms based on five-fold cross-validation. HGCNMDA achieves an AUC of 0.9626 and an average precision of 0.9660, respectively, which is ahead of other competitive algorithms. We further analyze the top-10 unknown interactions between miRNA and disease. In summary, HGCNMDA is a useful computational model for predicting miRNA-disease interactions.
Collapse
Affiliation(s)
- Chunyan Li
- School of Informatics, Xiamen University, Xiamen 361005, China
- Graduate School, Yunnan Minzu University, Kunming 650504, China
| | - Hongju Liu
- College of Information Technology and Computer Science, University of the Cordilleras, Baguio 2600, Philippines
| | - Qian Hu
- School of Informatics, Xiamen University, Xiamen 361005, China
| | - Jinlong Que
- School of Informatics, Xiamen University, Xiamen 361005, China
| | - Junfeng Yao
- School of Informatics, Xiamen University, Xiamen 361005, China.
| |
Collapse
|