1
|
Liu J, Wu R, Yuan S, Kelleher R, Chen S, Chen R, Zhang T, Obaidi I, Sheridan H. Pharmacogenomic Analysis of Combined Therapies against Glioblastoma Based on Cell Markers from Single-Cell Sequencing. Pharmaceuticals (Basel) 2023; 16:1533. [PMID: 38004399 PMCID: PMC10675611 DOI: 10.3390/ph16111533] [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: 08/27/2023] [Revised: 10/01/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma is the most common and aggressive form of primary brain cancer and the lack of viable treatment options has created an urgency to develop novel treatments. Personalized or predictive medicine is still in its infancy stage at present. This research aimed to discover biomarkers to inform disease progression and to develop personalized prophylactic and therapeutic strategies by combining state-of-the-art technologies such as single-cell RNA sequencing, systems pharmacology, and a polypharmacological approach. As predicted in the pyroptosis-related gene (PRG) transcription factor (TF) microRNA (miRNA) regulatory network, TP53 was the hub gene in the pyroptosis process in glioblastoma (GBM). A LASSO Cox regression model of pyroptosis-related genes was built to accurately and conveniently predict the one-, two-, and three-year overall survival rates of GBM patients. The top-scoring five natural compounds were parthenolide, rutin, baeomycesic acid, luteolin, and kaempferol, which have NFKB inhibition, antioxidant, lipoxygenase inhibition, glucosidase inhibition, and estrogen receptor agonism properties, respectively. In contrast, the analysis of the cell-type-specific differential expression-related targets of natural compounds showed that the top five subtype cells targeted by natural compounds were endothelial cells, microglia/macrophages, oligodendrocytes, dendritic cells, and neutrophil cells. The current approach-using the pharmacogenomic analysis of combined therapies-serves as a model for novel personalized therapeutic strategies for GBM treatment.
Collapse
Affiliation(s)
- Junying Liu
- NatPro Center, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland; (T.Z.); (I.O.); (H.S.)
| | - Ruixin Wu
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 274, Zhijiang Road, Jing’an District, Shanghai 200071, China;
| | - Shouli Yuan
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China;
| | - Robbie Kelleher
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland;
| | - Siying Chen
- The Second Affiliated Hospital, Nanchang University, Nanchang 330031, China;
| | - Rongfeng Chen
- National Center for Occupational Safety and Health, NHC, Beijing 102308, China;
| | - Tao Zhang
- NatPro Center, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland; (T.Z.); (I.O.); (H.S.)
- School of Food Science & Environmental Health, Technological University Dublin, D07 EWV4 Dublin, Ireland
| | - Ismael Obaidi
- NatPro Center, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland; (T.Z.); (I.O.); (H.S.)
| | - Helen Sheridan
- NatPro Center, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland; (T.Z.); (I.O.); (H.S.)
| |
Collapse
|
2
|
CFGM: An algorithm for closed frequent graph patterns mining. Inf Sci (N Y) 2023. [DOI: 10.1016/j.ins.2022.12.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
3
|
Wang Z, Zhang Y, Li Q, Zou Q, Liu Q. A road map for happiness: The psychological factors related cell types in various parts of human body from single cell RNA-seq data analysis. Comput Biol Med 2022; 143:105286. [PMID: 35183972 DOI: 10.1016/j.compbiomed.2022.105286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022]
Abstract
Massive evidence from all sources including zoology, neurobiology and immunology has confirmed that psychological factors can raise remarkable physiological effects. Researchers have long been aware of the potential value of these effects and wanted to harness them in the development of new drugs and therapies, for which the mechanism study is a necessary prerequisite. However, most of these studies are restricted to neuroscience, or starts with blood sample and fall into the area of immunity. In this study, we choose to focus on the psychological factor of happiness, mining existing publicly available single cell RNA sequencing (scRNA-seq) data for the expression of happiness-related genes collected from various sources of literature in all types of cells in the samples, finding that the expression of these genes is not restricted within neuro-regulated cells or tissue-resident immune cells, on the opposite, cell types that are unique to tissue and organ without direct regulation from nervous system account for the majority to express the happiness-related genes. Our research is a preliminary exploration of where our body respond to our mind at cell level, and lays the foundation for more detailed mechanism research.
Collapse
Affiliation(s)
- Ziwei Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology, China
| | - Ying Zhang
- Department of Anesthesiology, Hospital T.C.M Affiliated to Southwest Medical University, Luzhou, China
| | - Qun Li
- Department of Pain, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology, China; Yangtze Delta Region Institute Quzhou, University of Electronic Science and Technology of China, Quzhou, Zhejiang, China.
| | - Qing Liu
- Department of Algology, Hospital T.C.M Affiliated to Southwest Medical University, Luzhou, China.
| |
Collapse
|
4
|
Nussinov R, Tsai CJ, Jang H. How can same-gene mutations promote both cancer and developmental disorders? SCIENCE ADVANCES 2022; 8:eabm2059. [PMID: 35030014 PMCID: PMC8759737 DOI: 10.1126/sciadv.abm2059] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/22/2021] [Indexed: 05/05/2023]
Abstract
The question of how same-gene mutations can drive both cancer and neurodevelopmental disorders has been puzzling. It has also been puzzling why those with neurodevelopmental disorders have a high risk of cancer. Ras, MEK, PI3K, PTEN, and SHP2 are among the oncogenic proteins that can harbor mutations that encode diseases other than cancer. Understanding why some of their mutations can promote cancer, whereas others promote neurodevelopmental diseases, and why even the same mutations may promote both phenotypes, has important clinical ramifications. Here, we review the literature and address these tantalizing questions. We propose that cell type–specific expression of the mutant protein, and of other proteins in the respective pathway, timing of activation (during embryonic development or sporadic emergence), and the absolute number of molecules that the mutations activate, alone or in combination, are pivotal in determining the pathological phenotypes—cancer and (or) developmental disorders.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
5
|
Liu J, Wang H, Sun W, Liu Y. Prioritizing Autism Risk Genes using Personalized Graphical Models Estimated from Single Cell RNA-seq Data. J Am Stat Assoc 2022; 117:38-51. [PMID: 35529781 PMCID: PMC9070996 DOI: 10.1080/01621459.2021.1933495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hundreds of autism risk genes have been reported recently, mainly based on genetic studies where these risk genes have more de novo mutations in autism subjects than healthy controls. However, as a complex disease, autism is likely associated with more risk genes and many of them may not be identifiable through de novo mutations. We hypothesize that more autism risk genes can be identified through their connections with known autism risk genes in personalized gene-gene interaction graphs. We estimate such personalized graphs using single cell RNA sequencing (scRNA-seq) while appropriately modeling the cell dependence and possible zero-inflation in the scRNA-seq data. The sample size, which is the number of cells per individual, ranges from 891 to 1,241 in our case study using scRNA-seq data in autism subjects and controls. We consider 1,500 genes in our analysis. Since the number of genes is larger or comparable to the sample size, we perform penalized estimation. We score each gene's relevance by applying a simple graph kernel smoothing method to each personalized graph. The molecular functions of the top-scored genes are related to autism diseases. For example, a candidate gene RYR2 that encodes protein ryanodine receptor 2 is involved in neurotransmission, a process that is impaired in ASD patients. While our method provides a systemic and unbiased approach to prioritize autism risk genes, the relevance of these genes needs to be further validated in functional studies.
Collapse
Affiliation(s)
- Jianyu Liu
- Department of Statistics and Operations Research, University of North Carolina, Chapel Hill
| | - Haodong Wang
- Department of Statistics and Operations Research, University of North Carolina, Chapel Hill
| | - Wei Sun
- Biostatistics Program, Public Health Sciences Division Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Yufeng Liu
- Department of Statistics and Operations Research, University of North Carolina, Chapel Hill,Department of Genetics, Department of Biostatistics, Carolina Center for Genome Science, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill,
| |
Collapse
|
6
|
Chen X, Lin Y, Qu Q, Ning B, Chen H, Li X. An epistasis and heterogeneity analysis method based on maximum correlation and maximum consistence criteria. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:7711-7726. [PMID: 34814271 DOI: 10.3934/mbe.2021382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Tumor heterogeneity significantly increases the difficulty of tumor treatment. The same drugs and treatment methods have different effects on different tumor subtypes. Therefore, tumor heterogeneity is one of the main sources of poor prognosis, recurrence and metastasis. At present, there have been some computational methods to study tumor heterogeneity from the level of genome, transcriptome, and histology, but these methods still have certain limitations. In this study, we proposed an epistasis and heterogeneity analysis method based on genomic single nucleotide polymorphism (SNP) data. First of all, a maximum correlation and maximum consistence criteria was designed based on Bayesian network score K2 and information entropy for evaluating genomic epistasis. As the number of SNPs increases, the epistasis combination space increases sharply, resulting in a combination explosion phenomenon. Therefore, we next use an improved genetic algorithm to search the SNP epistatic combination space for identifying potential feasible epistasis solutions. Multiple epistasis solutions represent different pathogenic gene combinations, which may lead to different tumor subtypes, that is, heterogeneity. Finally, the XGBoost classifier is trained with feature SNPs selected that constitute multiple sets of epistatic solutions to verify that considering tumor heterogeneity is beneficial to improve the accuracy of tumor subtype prediction. In order to demonstrate the effectiveness of our method, the power of multiple epistatic recognition and the accuracy of tumor subtype classification measures are evaluated. Extensive simulation results show that our method has better power and prediction accuracy than previous methods.
Collapse
Affiliation(s)
- Xia Chen
- School of Basic Education, Changsha Aeronautical Vocational and Technical College, Changsha, Hunan 410124, China
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yexiong Lin
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Qiang Qu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Bin Ning
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Haowen Chen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Xiong Li
- School of Software, East China Jiaotong University, Nanchang 330013, China
| |
Collapse
|
7
|
Kosvyra A, Ntzioni E, Chouvarda I. Network analysis with biological data of cancer patients: A scoping review. J Biomed Inform 2021; 120:103873. [PMID: 34298154 DOI: 10.1016/j.jbi.2021.103873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/30/2021] [Accepted: 07/18/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND & OBJECTIVE Network Analysis (NA) is a mathematical method that allows exploring relations between units and representing them as a graph. Although NA was initially related to social sciences, the past two decades was introduced in Bioinformatics. The recent growth of the networks' use in biological data analysis reveals the need to further investigate this area. In this work, we attempt to identify the use of NA with biological data, and specifically: (a) what types of data are used and whether they are integrated or not, (b) what is the purpose of this analysis, predictive or descriptive, and (c) the outcome of such analyses, specifically in cancer diseases. METHODS & MATERIALS The literature review was conducted on two databases, PubMed & IEEE, and was restricted to journal articles of the last decade (January 2010 - December 2019). At a first level, all articles were screened by title and abstract, and at a second level the screening was conducted by reading the full text article, following the predefined inclusion & exclusion criteria leading to 131 articles of interest. A table was created with the information of interest and was used for the classification of the articles. The articles were initially classified to analysis studies and studies that propose a new algorithm or methodology. Each one of these categories was further screened by the following clustering criteria: (a) data used, (b) study purpose, (c) study outcome. Specifically for the studies proposing a new algorithm, the novelty presented in each one was detected. RESULTS & Conclusions: In the past five years researchers are focusing on creating new algorithms and methodologies to enhance this field. The articles' classification revealed that only 25% of the analyses are integrating multi-omics data, although 50% of the new algorithms developed follow this integrative direction. Moreover, only 20% of the analyses and 10% of the newly developed methodologies have a predictive purpose. Regarding the result of the works reviewed, 75% of the studies focus on identifying, prognostic or not, gene signatures. Concluding, this review revealed the need for deploying predictive and multi-omics integrative algorithms and methodologies that can be used to enhance cancer diagnosis, prognosis and treatment.
Collapse
Affiliation(s)
- A Kosvyra
- Laboratory of Computing, Medical Informatics and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - E Ntzioni
- Laboratory of Computing, Medical Informatics and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - I Chouvarda
- Laboratory of Computing, Medical Informatics and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
8
|
Wu Z, Lawrence PJ, Ma A, Zhu J, Xu D, Ma Q. Single-Cell Techniques and Deep Learning in Predicting Drug Response. Trends Pharmacol Sci 2020; 41:1050-1065. [PMID: 33153777 PMCID: PMC7669610 DOI: 10.1016/j.tips.2020.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022]
Abstract
Rapidly developing single-cell sequencing analyses produce more comprehensive profiles of the genomic, transcriptomic, and epigenomic heterogeneity of tumor subpopulations than do traditional bulk sequencing analyses. Moreover, single-cell techniques allow the response of a tumor to drug exposure to be more thoroughlyinvestigated. Deep learning (DL) models have successfully extracted features from complex bulk sequence data to predict drug responses. We review recent innovations in single-cell technologies and DL-based approaches related to drug sensitivity predictions. We believe that, by using insights from bulk sequencedata, deep transfer learning (DTL) can facilitate the use of single-cell data for training superior DL-based drug prediction models.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Patrick J Lawrence
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Jian Zhu
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
9
|
Li Y, Ma A, Mathé EA, Li L, Liu B, Ma Q. Elucidation of Biological Networks across Complex Diseases Using Single-Cell Omics. Trends Genet 2020; 36:951-966. [PMID: 32868128 DOI: 10.1016/j.tig.2020.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022]
Abstract
Single-cell multimodal omics (scMulti-omics) technologies have made it possible to trace cellular lineages during differentiation and to identify new cell types in heterogeneous cell populations. The derived information is especially promising for computing cell-type-specific biological networks encoded in complex diseases and improving our understanding of the underlying gene regulatory mechanisms. The integration of these networks could, therefore, give rise to a heterogeneous regulatory landscape (HRL) in support of disease diagnosis and drug therapeutics. In this review, we provide an overview of this field and pay particular attention to how diverse biological networks can be inferred in a specific cell type based on integrative methods. Then, we discuss how HRL can advance our understanding of regulatory mechanisms underlying complex diseases and aid in the prediction of prognosis and therapeutic responses. Finally, we outline challenges and future trends that will be central to bringing the field of HRL in complex diseases forward.
Collapse
Affiliation(s)
- Yang Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Ewy A Mathé
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Rockville, MD, 20892, USA
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China.
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Hie B, Peters J, Nyquist SK, Shalek AK, Berger B, Bryson BD. Computational Methods for Single-Cell RNA Sequencing. Annu Rev Biomed Data Sci 2020. [DOI: 10.1146/annurev-biodatasci-012220-100601] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Single-cell RNA sequencing (scRNA-seq) has provided a high-dimensional catalog of millions of cells across species and diseases. These data have spurred the development of hundreds of computational tools to derive novel biological insights. Here, we outline the components of scRNA-seq analytical pipelines and the computational methods that underlie these steps. We describe available methods, highlight well-executed benchmarking studies, and identify opportunities for additional benchmarking studies and computational methods. As the biochemical approaches for single-cell omics advance, we propose coupled development of robust analytical pipelines suited for the challenges that new data present and principled selection of analytical methods that are suited for the biological questions to be addressed.
Collapse
Affiliation(s)
- Brian Hie
- Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Joshua Peters
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
| | - Sarah K. Nyquist
- Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Alex K. Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
- Department of Chemistry, Institute for Medical Engineering & Science (IMES), and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Bryan D. Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
11
|
Peng L, Tian X, Tian G, Xu J, Huang X, Weng Y, Yang J, Zhou L. Single-cell RNA-seq clustering: datasets, models, and algorithms. RNA Biol 2020; 17:765-783. [PMID: 32116127 PMCID: PMC7549635 DOI: 10.1080/15476286.2020.1728961] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/13/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technologies allow numerous opportunities for revealing novel and potentially unexpected biological discoveries. scRNA-seq clustering helps elucidate cell-to-cell heterogeneity and uncover cell subgroups and cell dynamics at the group level. Two important aspects of scRNA-seq data analysis were introduced and discussed in the present review: relevant datasets and analytical tools. In particular, we reviewed popular scRNA-seq datasets and discussed scRNA-seq clustering models including K-means clustering, hierarchical clustering, consensus clustering, and so on. Seven state-of-the-art scRNA clustering methods were compared on five public available datasets. Two primary evaluation metrics, the Adjusted Rand Index (ARI) and the Normalized Mutual Information (NMI), were used to evaluate these methods. Although unsupervised models can effectively cluster scRNA-seq data, these methods also have challenges. Some suggestions were provided for future research directions.
Collapse
Affiliation(s)
- Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Xiongfei Tian
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Geng Tian
- Geneis (Beijing) Co. Ltd, Beijing, China
| | - Junlin Xu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xin Huang
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Yanbin Weng
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | | | - Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| |
Collapse
|