1
|
Liu H, Lin S, Chen PX, Min J, Liu XY, Guan T, Yang CY, Xiao XJ, Xiong DH, Sun SJ, Nie L, Gong H, Wu XS, He XF, Liu J. Integrated bioinformatics analysis to develop diagnostic models for malignant transformation of chronic proliferative diseases. BLOOD SCIENCE 2025; 7:e00226. [PMID: 40201199 PMCID: PMC11977743 DOI: 10.1097/bs9.0000000000000226] [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: 11/05/2024] [Accepted: 02/16/2025] [Indexed: 04/10/2025] Open
Abstract
The combined analysis of dual diseases can provide new insights into pathogenic mechanisms, identify novel biomarkers, and develop targeted therapeutic strategies. Polycythemia vera (PV) is a chronic myeloproliferative neoplasm associated with a risk of acute myeloid leukemia (AML) transformation. However, the chronic nature of disease transformation complicates longitudinal high-throughput sequencing studies of patients with PV before and after AML transformation. This study aimed to develop a diagnostic model for malignant transformation of chronic proliferative diseases, addressing the challenges of early detection and intervention. Integrated public datasets of PV and AML were analyzed to identify differentially expressed genes (DEGs) and construct a weighted correlation network. Machine-learning algorithms screen genes for potential biomarkers, leading to the development of diagnostic models. Clinical specimens were collected to validate gene expression. cMAP and molecular docking predicted potential drugs. In vitro experiments were performed to assess drug efficacy in PV and AML cells. CIBERSORT and single-cell RNA-sequencing (scRNA-seq) analyses were used to explore the impact of hub genes on the tumor microenvironment. We identified 24 genes shared between PV and AML, which were enriched in immune-related pathways. Lactoferrin (LTF) and G protein-coupled receptor 65 (GPR65) were integrated into a nomogram with a robust predictive power. The predicted drug vemurafenib inhibited proliferation and increased apoptosis in PV and AML cells. TME analysis has linked these biomarkers to macrophages. Clinical samples were used to confirm LTF and GPR65 expression levels. We identified shared genes between PV and AML and developed a diagnostic nomogram that offers a novel avenue for the diagnosis and clinical management of AML-related PV.
Collapse
Affiliation(s)
- Hua Liu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Sheng Lin
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Pei-Xuan Chen
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Juan Min
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xia-Yang Liu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Ting Guan
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Chao-Ying Yang
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xiao-Juan Xiao
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - De-Hui Xiong
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Sheng-Jie Sun
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Ling Nie
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Han Gong
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xu-Sheng Wu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xiao-Feng He
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Jing Liu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| |
Collapse
|
2
|
Yan J, Yang Y, Liu Y, Shi X, Wu H, Dai M. MicroRNA let-7g links foam cell formation and adipogenic differentiation: A key regulator of Paeonol treating atherosclerosis-osteoporosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155447. [PMID: 38394732 DOI: 10.1016/j.phymed.2024.155447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/30/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUD High comorbidity rates have been reported in patients with atherosclerosis and osteoporosis, posing a serious risk to the health and well-being of elderly patients. To improve and update clinical practice regarding the joint treatment of these two diseases, the common mechanisms of atherosclerosis and osteoporosis need to be clarified. MicroRNAs (miRNAs), are importance molecules in the pathogenesis of human diseases, including in cardiovascular and orthopedic fields. They have garnered interest as potential targets for novel therapeutic strategies. However, the key miRNAs involved in atherosclerosis and osteoporosis and their precise regulation mechanisms remain unknown. Paeonol (Pae), an active ingredient in Cortex Moutan, has shown promising results in improving both lipid and bone metabolic abnormalities. However, it is uncertain whether this agent can exert a cotherapeutic effect on atherosclerosis and osteoporosis. OBJECTIVE This study aimed to screen important shared miRNAs in atherosclerotic and osteoporotic complications, and explore the mechanism of the protective effects of Pae against atherosclerosis and osteoporosis in high-fat diet (HFD)-fed ApoE-/- mice. METHODS An experimental atherosclerosis and osteoporosis model was established in 40-week-old HFD ApoE-/- mice. Various techniques such as Oil Red O staining, HE staining and micro-CT were used to confirm the co-occurrence of these two diseases and efficacy of Pae in addition to the associated biochemical changes. Bioinformatics was used to screen key miRNAs in the atherosclerosis and osteoporosis model, and gene involvement was assessed through serum analyses, qRT-PCR, and western blot. To investigate the effect of Pae on the modulation of the miR let-7g/HMGA2/CEBPβ pathway, Raw 264.7 cells were cocultured with bone marrow mesenchymal stem cells (BMSCs) and treated with an miR let-7g mimic/inhibitor. RESULTS miR let-7g identified using bioinformatics was assessed to evaluate its participation in atherosclerosis-osteoporosis. Experimental analysis showed reduced miR let-7g levels in the atherosclerosis-osteoporosis mice model. Moreover, miR let-7g was required for BMSC - Raw 264.7 cell crosstalk, thereby promoting foam cell formation and adipocyte differentiation. Treatment with Pae significantly reduced plaque accumulation and foam cell number in the aorta while increasing bone density and improving trabecular bone microarchitecture in HFD ApoE-/- mice. Pae also increased the level of miR let-7g in the bloodstream of model mice. In vitro studies, Pae enhanced miR let-7g expression in BMSCs, thereby suppressing the HMGA2/CEBPβ pathway to prevent the formation of foam cells and differentiation of adipocytes induced by oxidized low-density lipoprotein (ox-LDL). CONCLUSION The study results suggested that miR let-7g participates in atherosclerosis -osteoporosis regulation and that Pae acts as a potential therapeutic agent for preventing atherosclerosis-osteoporosis through regulatory effects on the miR let-7g/HMGA2/CEBPβ pathway to hinder foam cell formation and adipocyte differentiation.
Collapse
Affiliation(s)
- Jinjin Yan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Yulong Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Yarong Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Xiaoyan Shi
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Hongfei Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, PR China.
| | - Min Dai
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, PR China.
| |
Collapse
|
3
|
Liu P, Luo J, Chen X. miRCom: Tensor Completion Integrating Multi-View Information to Deduce the Potential Disease-Related miRNA-miRNA Pairs. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1747-1759. [PMID: 33180730 DOI: 10.1109/tcbb.2020.3037331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
MicroRNAs (miRNAs) are consistently capable of regulating gene expression synergistically in a combination mode and play a key role in various biological processes associated with the initiation and development of human diseases, which indicate that comprehending the synergistic molecular mechanism of miRNAs may facilitate understanding the pathogenesis of diseases or even overcome it. However, most existing computational methods had an incomprehensive acknowledge of the miRNA synergistic effect on the pathogenesis of complex diseases, or were hard to be extended to a large-scale prediction task of miRNA synergistic combinations for different diseases. In this article, we propose a novel tensor completion framework integrating multi-view miRNAs and diseases information, called miRCom, for the discovery of potential disease-associated miRNA-miRNA pairs. We first construct an incomplete three-order association tensor and several types of similarity matrices based on existing biological knowledge. Then, we formulate an objective function via performing the factorizations of coupled tensor and matrices simultaneously. Finally, we build an optimization schema by adopting the ADMM algorithm. After that, we obtain the prediction of miRNA-miRNA pairs for different diseases from the full tensor. The contrastive experimental results with other approaches verified that miRCom effectively identify the potential disease-related miRNA-miRNA pairs. Moreover, case study results further illustrated that miRNA-miRNA pairs have more biologically significance and prognostic value than single miRNAs.
Collapse
|
4
|
Pesaranghader A, Matwin S, Sokolova M, Grenier JC, Beiko RG, Hussin J. OUP accepted manuscript. Bioinformatics 2022; 38:3051-3061. [PMID: 35536192 PMCID: PMC9154256 DOI: 10.1093/bioinformatics/btac304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/12/2022] [Indexed: 11/24/2022] Open
Abstract
Motivation There is a plethora of measures to evaluate functional similarity (FS) of genes based on their co-expression, protein–protein interactions and sequence similarity. These measures are typically derived from hand-engineered and application-specific metrics to quantify the degree of shared information between two genes using their Gene Ontology (GO) annotations. Results We introduce deepSimDEF, a deep learning method to automatically learn FS estimation of gene pairs given a set of genes and their GO annotations. deepSimDEF’s key novelty is its ability to learn low-dimensional embedding vector representations of GO terms and gene products and then calculate FS using these learned vectors. We show that deepSimDEF can predict the FS of new genes using their annotations: it outperformed all other FS measures by >5–10% on yeast and human reference datasets on protein–protein interactions, gene co-expression and sequence homology tasks. Thus, deepSimDEF offers a powerful and adaptable deep neural architecture that can benefit a wide range of problems in genomics and proteomics, and its architecture is flexible enough to support its extension to any organism. Availability and implementation Source code and data are available at https://github.com/ahmadpgh/deepSimDEF Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Stan Matwin
- Faculty of Computer Science, Dalhousie University, Halifax B3H 4R2, Canada
- Institute for Big Data Analytics, Dalhousie University, Halifax B3H 4R2, Canada
- Institute of Computer Science, Polish Academy of Sciences, Warsaw, Poland
| | - Marina Sokolova
- Institute for Big Data Analytics, Dalhousie University, Halifax B3H 4R2, Canada
- Faculty of Medicine and Faculty of Engineering, University of Ottawa, Ottawa K1H 8M5, Canada
| | | | - Robert G Beiko
- Faculty of Computer Science, Dalhousie University, Halifax B3H 4R2, Canada
- Institute for Big Data Analytics, Dalhousie University, Halifax B3H 4R2, Canada
| | | |
Collapse
|
5
|
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
|
6
|
Yi HC, You ZH, Huang DS, Kwoh CK. Graph representation learning in bioinformatics: trends, methods and applications. Brief Bioinform 2021; 23:6361044. [PMID: 34471921 DOI: 10.1093/bib/bbab340] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/18/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Graph is a natural data structure for describing complex systems, which contains a set of objects and relationships. Ubiquitous real-life biomedical problems can be modeled as graph analytics tasks. Machine learning, especially deep learning, succeeds in vast bioinformatics scenarios with data represented in Euclidean domain. However, rich relational information between biological elements is retained in the non-Euclidean biomedical graphs, which is not learning friendly to classic machine learning methods. Graph representation learning aims to embed graph into a low-dimensional space while preserving graph topology and node properties. It bridges biomedical graphs and modern machine learning methods and has recently raised widespread interest in both machine learning and bioinformatics communities. In this work, we summarize the advances of graph representation learning and its representative applications in bioinformatics. To provide a comprehensive and structured analysis and perspective, we first categorize and analyze both graph embedding methods (homogeneous graph embedding, heterogeneous graph embedding, attribute graph embedding) and graph neural networks. Furthermore, we summarize their representative applications from molecular level to genomics, pharmaceutical and healthcare systems level. Moreover, we provide open resource platforms and libraries for implementing these graph representation learning methods and discuss the challenges and opportunities of graph representation learning in bioinformatics. This work provides a comprehensive survey of emerging graph representation learning algorithms and their applications in bioinformatics. It is anticipated that it could bring valuable insights for researchers to contribute their knowledge to graph representation learning and future-oriented bioinformatics studies.
Collapse
Affiliation(s)
- Hai-Cheng Yi
- Chinese Academy of Sciences, Xinjiang Technical Institute of Physics and Chemistry, Urumqi 830011, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China
| | - De-Shuang Huang
- Institute of Machine Learning and Systems Biology, School of Electronics and Information Engineering, Tongji University, Shanghai 201804, China
| | - Chee Keong Kwoh
- School of Computer Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore
| |
Collapse
|
7
|
Chu Y, Wang X, Dai Q, Wang Y, Wang Q, Peng S, Wei X, Qiu J, Salahub DR, Xiong Y, Wei DQ. MDA-GCNFTG: identifying miRNA-disease associations based on graph convolutional networks via graph sampling through the feature and topology graph. Brief Bioinform 2021; 22:6261915. [PMID: 34009265 DOI: 10.1093/bib/bbab165] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
Accurate identification of the miRNA-disease associations (MDAs) helps to understand the etiology and mechanisms of various diseases. However, the experimental methods are costly and time-consuming. Thus, it is urgent to develop computational methods towards the prediction of MDAs. Based on the graph theory, the MDA prediction is regarded as a node classification task in the present study. To solve this task, we propose a novel method MDA-GCNFTG, which predicts MDAs based on Graph Convolutional Networks (GCNs) via graph sampling through the Feature and Topology Graph to improve the training efficiency and accuracy. This method models both the potential connections of feature space and the structural relationships of MDA data. The nodes of the graphs are represented by the disease semantic similarity, miRNA functional similarity and Gaussian interaction profile kernel similarity. Moreover, we considered six tasks simultaneously on the MDA prediction problem at the first time, which ensure that under both balanced and unbalanced sample distribution, MDA-GCNFTG can predict not only new MDAs but also new diseases without known related miRNAs and new miRNAs without known related diseases. The results of 5-fold cross-validation show that the MDA-GCNFTG method has achieved satisfactory performance on all six tasks and is significantly superior to the classic machine learning methods and the state-of-the-art MDA prediction methods. Moreover, the effectiveness of GCNs via the graph sampling strategy and the feature and topology graph in MDA-GCNFTG has also been demonstrated. More importantly, case studies for two diseases and three miRNAs are conducted and achieved satisfactory performance.
Collapse
Affiliation(s)
- Yanyi Chu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, China
| | - Xuhong Wang
- School of Electronic, Information and Electrical Engineering (SEIEE), Shanghai Jiao Tong University, China
| | - Qiuying Dai
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, China
| | - Yanjing Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, China
| | - Qiankun Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, China
| | | | | | - Dennis Russell Salahub
- Department of Chemistry, University of Calgary, Fellow Royal Society of Canada and Fellow of the American Association for the Advancement of Science, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
8
|
A Novel Method for Predicting Disease-Associated LncRNA-MiRNA Pairs Based on the Higher-Order Orthogonal Iteration. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2019; 2019:7614850. [PMID: 31191710 PMCID: PMC6525924 DOI: 10.1155/2019/7614850] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/25/2019] [Accepted: 02/10/2019] [Indexed: 12/30/2022]
Abstract
A lot of research studies have shown that many complex human diseases are associated not only with microRNAs (miRNAs) but also with long noncoding RNAs (lncRNAs). However, most of the current existing studies focus on the prediction of disease-related miRNAs or lncRNAs, and to our knowledge, until now, there are few literature studies reported to pay attention to the study of impact of miRNA-lncRNA pairs on diseases, although more and more studies have shown that both lncRNAs and miRNAs play important roles in cell proliferation and differentiation during the recent years. The identification of disease-related genes provides great insight into the underlying pathogenesis of diseases at a system level. In this study, a novel model called PADLMHOOI was proposed to predict potential associations between diseases and lncRNA-miRNA pairs based on the higher-order orthogonal iteration, and in order to evaluate its prediction performance, the global and local LOOCV were implemented, respectively, and simulation results demonstrated that PADLMHOOI could achieve reliable AUCs of 0.9545 and 0.8874 in global and local LOOCV separately. Moreover, case studies further demonstrated the effectiveness of PADLMHOOI to infer unknown disease-related lncRNA-miRNA pairs.
Collapse
|
9
|
Kuang L, Zhao H, Wang L, Xuan Z, Pei T. A Novel Approach Based on Point Cut Set to Predict Associations of Diseases and LncRNAs. Curr Bioinform 2019. [DOI: 10.2174/1574893613666181026122045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
In recent years, more evidence have progressively indicated that Long
non-coding RNAs (lncRNAs) play vital roles in wide-ranging human diseases, which can serve as
potential biomarkers and drug targets. Comparing with vast lncRNAs being found, the relationships
between lncRNAs and diseases remain largely unknown.
Objective:
The prediction of novel and potential associations between lncRNAs and diseases would
contribute to dissect the complex mechanisms of disease pathogenesis.
associations while known disease-lncRNA associations are required only.
Method:
In this paper, a new computational method based on Point Cut Set is proposed to predict
LncRNA-Disease Associations (PCSLDA) based on known lncRNA-disease associations. Compared
with the existing state-of-the-art methods, the major novelty of PCSLDA lies in the incorporation of
distance difference matrix and point cut set to set the distance correlation coefficient of nodes in the
lncRNA-disease interaction network. Hence, PCSLDA can be applied to forecast potential lncRNAdisease
associations while known disease-lncRNA associations are required only.
Results:
Simulation results show that PCSLDA can significantly outperform previous state-of-the-art
methods with reliable AUC of 0.8902 in the leave-one-out cross-validation and AUCs of 0.7634 and
0.8317 in 5-fold cross-validation and 10-fold cross-validation respectively. And additionally, 70% of
top 10 predicted cancer-lncRNA associations can be confirmed.
Conclusion:
It is anticipated that our proposed model can be a great addition to the biomedical
research field.
Collapse
Affiliation(s)
- Linai Kuang
- Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, Xiangtan, China
| | - Haochen Zhao
- Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, Xiangtan, China
| | - Lei Wang
- Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, Xiangtan, China
| | - Zhanwei Xuan
- Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, Xiangtan, China
| | - Tingrui Pei
- Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, Xiangtan, China
| |
Collapse
|
10
|
Ping P, Wang L, Kuang L, Ye S, Iqbal MFB, Pei T. A Novel Method for LncRNA-Disease Association Prediction Based on an lncRNA-Disease Association Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:688-693. [PMID: 29993639 DOI: 10.1109/tcbb.2018.2827373] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An increasing number of studies have indicated that long-non-coding RNAs (lncRNAs) play critical roles in many important biological processes. Predicting potential lncRNA-disease associations can improve our understanding of the molecular mechanisms of human diseases and aid in finding biomarkers for disease diagnosis, treatment, and prevention. In this paper, we constructed a bipartite network based on known lncRNA-disease associations; based on this work, we proposed a novel model for inferring potential lncRNA-disease associations. Specifically, we analyzed the properties of the bipartite network and found that it closely followed a power-law distribution. Moreover, to evaluate the performance of our model, a leave-one-out cross-validation (LOOCV) framework was implemented, and the simulation results showed that our computational model significantly outperformed previous state-of-the-art models, with AUCs of 0.8825, 0.9004, and 0.9292 for known lncRNA-disease associations obtained from the LncRNADisease database, Lnc2Cancer database, and MNDR database, respectively. Thus, our approach may be an excellent addition to the biomedical research field in the future.
Collapse
|
11
|
A Novel Model for Predicting Associations between Diseases and LncRNA-miRNA Pairs Based on a Newly Constructed Bipartite Network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2018; 2018:6789089. [PMID: 29853986 PMCID: PMC5960578 DOI: 10.1155/2018/6789089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/16/2018] [Accepted: 03/26/2018] [Indexed: 11/18/2022]
Abstract
Motivation Increasing studies have demonstrated that many human complex diseases are associated with not only microRNAs, but also long-noncoding RNAs (lncRNAs). LncRNAs and microRNA play significant roles in various biological processes. Therefore, developing effective computational models for predicting novel associations between diseases and lncRNA-miRNA pairs (LMPairs) will be beneficial to not only the understanding of disease mechanisms at lncRNA-miRNA level and the detection of disease biomarkers for disease diagnosis, treatment, prognosis, and prevention, but also the understanding of interactions between diseases and LMPairs at disease level. Results It is well known that genes with similar functions are often associated with similar diseases. In this article, a novel model named PADLMP for predicting associations between diseases and LMPairs is proposed. In this model, a Disease-LncRNA-miRNA (DLM) tripartite network was designed firstly by integrating the lncRNA-disease association network and miRNA-disease association network; then we constructed the disease-LMPairs bipartite association network based on the DLM network and lncRNA-miRNA association network; finally, we predicted potential associations between diseases and LMPairs based on the newly constructed disease-LMPair network. Simulation results show that PADLMP can achieve AUCs of 0.9318, 0.9090 ± 0.0264, and 0.8950 ± 0.0027 in the LOOCV, 2-fold, and 5-fold cross validation framework, respectively, which demonstrate the reliable prediction performance of PADLMP.
Collapse
|
12
|
Zhao H, Kuang L, Wang L, Ping P, Xuan Z, Pei T, Wu Z. Prediction of microRNA-disease associations based on distance correlation set. BMC Bioinformatics 2018; 19:141. [PMID: 29665774 PMCID: PMC5905221 DOI: 10.1186/s12859-018-2146-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/03/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Recently, numerous laboratory studies have indicated that many microRNAs (miRNAs) are involved in and associated with human diseases and can serve as potential biomarkers and drug targets. Therefore, developing effective computational models for the prediction of novel associations between diseases and miRNAs could be beneficial for achieving an understanding of disease mechanisms at the miRNA level and the interactions between diseases and miRNAs at the disease level. Thus far, only a few miRNA-disease association pairs are known, and models analyzing miRNA-disease associations based on lncRNA are limited. RESULTS In this study, a new computational method based on a distance correlation set is developed to predict miRNA-disease associations (DCSMDA) by integrating known lncRNA-disease associations, known miRNA-lncRNA associations, disease semantic similarity, and various lncRNA and disease similarity measures. The novelty of DCSMDA is due to the construction of a miRNA-lncRNA-disease network, which reveals that DCSMDA can be applied to predict potential lncRNA-disease associations without requiring any known miRNA-disease associations. Although the implementation of DCSMDA does not require known disease-miRNA associations, the area under curve is 0.8155 in the leave-one-out cross validation. Furthermore, DCSMDA was implemented in case studies of prostatic neoplasms, lung neoplasms and leukaemia, and of the top 10 predicted associations, 10, 9 and 9 associations, respectively, were separately verified in other independent studies and biological experimental studies. In addition, 10 of the 10 (100%) associations predicted by DCSMDA were supported by recent bioinformatical studies. CONCLUSIONS According to the simulation results, DCSMDA can be a great addition to the biomedical research field.
Collapse
Affiliation(s)
- Haochen Zhao
- Key Laboratory of Intelligent Computing & Information Processing (Xiangtan University), Ministry of Education, China, Xiangtan, 411105, Hunan, People's Republic of China.,College of Information Engineering, Xiangtan University, Xiangtan, Hunan, People's Republic of China
| | - Linai Kuang
- College of Computer Engineering & Applied Mathematics, Changsha University, Changsha, 410001, Hunan, People's Republic of China.,Key Laboratory of Intelligent Computing & Information Processing (Xiangtan University), Ministry of Education, China, Xiangtan, 411105, Hunan, People's Republic of China.,College of Information Engineering, Xiangtan University, Xiangtan, Hunan, People's Republic of China
| | - Lei Wang
- College of Computer Engineering & Applied Mathematics, Changsha University, Changsha, 410001, Hunan, People's Republic of China. .,Key Laboratory of Intelligent Computing & Information Processing (Xiangtan University), Ministry of Education, China, Xiangtan, 411105, Hunan, People's Republic of China. .,Department of Computer Science, Lakehead University, Thunder Bay, ON, P7B5E1, Canada. .,College of Information Engineering, Xiangtan University, Xiangtan, Hunan, People's Republic of China.
| | - Pengyao Ping
- Key Laboratory of Intelligent Computing & Information Processing (Xiangtan University), Ministry of Education, China, Xiangtan, 411105, Hunan, People's Republic of China.,College of Information Engineering, Xiangtan University, Xiangtan, Hunan, People's Republic of China
| | - Zhanwei Xuan
- Key Laboratory of Intelligent Computing & Information Processing (Xiangtan University), Ministry of Education, China, Xiangtan, 411105, Hunan, People's Republic of China.,College of Information Engineering, Xiangtan University, Xiangtan, Hunan, People's Republic of China
| | - Tingrui Pei
- Key Laboratory of Intelligent Computing & Information Processing (Xiangtan University), Ministry of Education, China, Xiangtan, 411105, Hunan, People's Republic of China.,College of Information Engineering, Xiangtan University, Xiangtan, Hunan, People's Republic of China
| | - Zhelun Wu
- Department of Computer Science, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|