51
|
Ren L, Zhao D, Zhao X, Chen W, Li L, Wu T, Liang G, Cai Z, Xu S. Multi-level thresholding segmentation for pathological images: Optimal performance design of a new modified differential evolution. Comput Biol Med 2022; 148:105910. [DOI: 10.1016/j.compbiomed.2022.105910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/11/2022] [Accepted: 07/23/2022] [Indexed: 02/07/2023]
|
52
|
Hu Y, Tang C, Zhu W, Ye H, Lin Y, Wang R, Zhou T, Wen S, Yang J, Fang C. Identification of chromosomal instability-associated genes as hepatocellular carcinoma progression-related biomarkers to guide clinical diagnosis, prognosis and therapy. Comput Biol Med 2022; 148:105896. [PMID: 35868048 DOI: 10.1016/j.compbiomed.2022.105896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/21/2022] [Accepted: 07/16/2022] [Indexed: 11/03/2022]
Abstract
Hepatocellular carcinoma (HCC) is a type of cancer characterized by high heterogeneity and a complex multistep progression process. Significantly-altered biomarkers for HCC need to be identified. Differentially expressed genes and weighted gene co-expression network analyses were used to identify progression-related biomarkers. LASSO-Cox regression and random forest algorithms were used to construct the progression-related prognosis (PRP) score. Three chromosomal instability-associated genes (KIF20A, TOP2A, and TTK) have been identified as progression-related biomarkers. The robustness of the PRP scores were validated using four independent cohorts. Immune status was observed using the single-sample gene set enrichment analysis (ssGSEA). Comprehensive analysis showed that the patients with high PRP score had wider genomic alterations, more malignant phenotypes, and were in a state of immunosuppression. The diagnostic models constructed via logistic regression based on the three genes showed satisfactory performances in distinguishing HCC from cirrhotic tissues or dysplastic nodules. The nomogram combining PRP scores with clinical factors had a better performance in predicting prognosis than the tumor node metastasis classification (TNM) system. We further confirmed that KIF20A, TOP2A, and TTK were highly expressed in HCC tissues than in cirrhotic tissues. Downregulation of all three genes aggravated chromosomal instabilities in HCC and suppressed HCC cells viability both in vitro and in vivo. Overall, our study highlights the important roles of chromosomal instability-associated genes during the progression of HCC and their potential clinical diagnosis and prognostic value and provides promising new ideas for developing therapeutic strategies to improve the outcomes of HCC patients.
Collapse
Affiliation(s)
- Yueyang Hu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Chuanyu Tang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Wen Zhu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Hanjie Ye
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Yuxing Lin
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Ruixuan Wang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Tianjun Zhou
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Sai Wen
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Jian Yang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Institute of Digital Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China.
| |
Collapse
|
53
|
Predicting Entrepreneurial Intention of Students: Kernel Extreme Learning Machine with Boosted Crow Search Algorithm. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12146907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
College students are the group with the most entrepreneurial vitality and potential. How to cultivate their entrepreneurial and innovative ability is one of the important and urgent issues facing this current social development. This paper proposes a reliable, intelligent prediction model of entrepreneurial intentions, providing theoretical support for guiding college students’ positive entrepreneurial intentions. The model mainly uses the improved crow search algorithm (CSA) to optimize the kernel extreme learning machine (KELM) model with feature selection (FS), namely CSA-KELM-FS, to study entrepreneurial intention. To obtain the best fitting model and key features, the gradient search rule, local escaping operator, and levy flight mutation (GLL) mechanism are introduced to enhance the CSA (GLLCSA), and FS is used to extract the key features. To verify the performance of the proposed GLLCSA, it is compared with eight other state-of-the-art methods. Further, the GLLCSA-KELM-FS model and five other machine learning methods have been used to predict the entrepreneurial intentions of 842 students from the Wenzhou Vocational College in Zhejiang, China, in the past five years. The results show that the proposed model can correctly predict the students’ entrepreneurial intention with an accuracy rate of 93.2% and excellent stability. According to the prediction results of the proposed model, the key factors affecting the student’s entrepreneurial intention are mainly the major studied, campus innovation, entrepreneurship practice experience, and positive personality. Therefore, the proposed GLLCSA-KELM-FS is expected to be an effective tool for predicting students’ entrepreneurial intentions.
Collapse
|
54
|
Multilevel threshold image segmentation for COVID-19 chest radiography: A framework using horizontal and vertical multiverse optimization. Comput Biol Med 2022; 146:105618. [PMID: 35690477 PMCID: PMC9113963 DOI: 10.1016/j.compbiomed.2022.105618] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 11/28/2022]
Abstract
COVID-19 is currently raging worldwide, with more patients being diagnosed every day. It usually is diagnosed by examining pathological photographs of the patient's lungs. There is a lot of detailed and essential information on chest radiographs, but manual processing is not as efficient or accurate. As a result, how efficiently analyzing and processing chest radiography of COVID-19 patients is an important research direction to promote COVID-19 diagnosis. To improve the processing efficiency of COVID-19 chest films, a multilevel thresholding image segmentation (MTIS) method based on an enhanced multiverse optimizer (CCMVO) is proposed. CCMVO is improved from the original Multi-Verse Optimizer by introducing horizontal and vertical search mechanisms. It has a more assertive global search ability and can jump out of the local optimum in optimization. The CCMVO-based MTIS method can obtain higher quality segmentation results than HHO, SCA, and other forms and is less prone to stagnation during the segmentation process. To verify the performance of the proposed CCMVO algorithm, CCMVO is first compared with DE, MVO, and other algorithms by 30 benchmark functions; then, the proposed CCMVO is applied to image segmentation of COVID-19 chest radiography; finally, this paper verifies that the combination of MTIS and CCMVO is very successful with good segmentation results by using the Feature Similarity Index (FSIM), the Peak Signal to Noise Ratio (PSNR), and the Structural Similarity Index (SSIM). Therefore, this research can provide an effective segmentation method for a medical organization to process COVID-19 chest radiography and then help doctors diagnose coronavirus pneumonia (COVID-19).
Collapse
|
55
|
A multi-omics machine learning framework in predicting the survival of colorectal cancer patients. Comput Biol Med 2022; 146:105516. [DOI: 10.1016/j.compbiomed.2022.105516] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 12/18/2022]
|
56
|
Nag A, Banerjee R, Paul S, Kundu R. Curcumin inhibits spike protein of new SARS-CoV-2 variant of concern (VOC) Omicron, an in silico study. Comput Biol Med 2022; 146:105552. [PMID: 35508082 PMCID: PMC9044632 DOI: 10.1016/j.compbiomed.2022.105552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Omicron (B.1.1.529), a variant of SARS-CoV-2 is currently spreading globally as a dominant strain. Due to multiple mutations at its Spike protein, including 15 amino acid substitutions at the receptor binding domain (RBD), Omicron is a variant of concern (VOC) and capable of escaping vaccine generated immunity. So far, no specific treatment regime is suggested for this VOC. METHODS The three-dimensional structure of the Spike RBD domain of Omicron variant was constructed by incorporating 15 amino acid substitutions to the Native Spike (S) structure and structural changes were compared that of the Native S. Seven phytochemicals namely Allicin, Capsaicin, Cinnamaldehyde, Curcumin, Gingerol, Piperine, and Zingeberene were docked with Omicron S protein and Omicron S-hACE2 complex. Further, molecular dynamic simulation was performed between Crcumin and Omicron S protein to evaluate the structural stability of the complex in the physiological environment and compared with that of the control drug Chloroquine. RESULTS Curcumin, among seven phytochemicals, was found to have the most substantial inhibitory potential with Omicron S protein. Further, it was found that curcumin could disrupt the Omicron S-hACE2 complex. The molecular dynamic simulation demonstrated that Curcumin could form a stable structure with Omicron S in the physiological environment. CONCLUSION To conclude, Curcumin can be considered as a potential therapeutic agent against the highly infectious Omicron variant of SARS-CoV-2.
Collapse
Affiliation(s)
- Anish Nag
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, 560029, India,Corresponding author
| | - Ritesh Banerjee
- School of Biological and Environmental Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Subhabrata Paul
- School of Biotechnology, Presidency University, Canal Bank Rd, DG Block, Action Area 1D, New Town, West Bengal, 700156, India
| | - Rita Kundu
- Department of Botany, University of Calcutta, Kolkata, West Bengal, 700019, India
| |
Collapse
|
57
|
An integrated pan-cancer analysis of identifying biomarkers about the EGR family genes in human carcinomas. Comput Biol Med 2022; 148:105889. [DOI: 10.1016/j.compbiomed.2022.105889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/25/2022] [Accepted: 07/16/2022] [Indexed: 12/24/2022]
|
58
|
Boosted Sine Cosine Algorithm with Application to Medical Diagnosis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6215574. [PMID: 35785140 PMCID: PMC9242811 DOI: 10.1155/2022/6215574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/19/2022] [Accepted: 05/26/2022] [Indexed: 01/09/2023]
Abstract
The sine cosine algorithm (SCA) was proposed for solving optimization tasks, of which the way to obtain the optimal solution is mainly through the continuous iteration of the sine and cosine update formulas. However, SCA also faces low population diversity and stagnation of locally optimal solutions. Hence, we try to eliminate these problems by proposing an enhanced version of SCA, named ESCA_PSO. ESCA_PSO is proposed based on hybrid SCA and particle swarm optimization (PSO) by incorporating multiple mutation strategies into the original SCA_PSO. To validate the effect of ESCA_PSO in handling global optimization problems, ESCA_PSO was compared with quality algorithms on various types of benchmark functions. In addition, the proposed ESCA_PSO was employed to tune the best parameters of support vector machines for dealing with medical diagnosis tasks. The results prove the efficiency of the proposed algorithms in solving optimization problems.
Collapse
|
59
|
Boosted machine learning model for predicting intradialytic hypotension using serum biomarkers of nutrition. Comput Biol Med 2022; 147:105752. [DOI: 10.1016/j.compbiomed.2022.105752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/22/2022]
|
60
|
Feng G, Yao H, Li C, Liu R, Huang R, Fan X, Ge R, Miao Q. ME-ACP: Multi-view neural networks with ensemble model for identification of anticancer peptides. Comput Biol Med 2022; 145:105459. [DOI: 10.1016/j.compbiomed.2022.105459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/26/2022]
|
61
|
Tool for Predicting College Student Career Decisions: An Enhanced Support Vector Machine Framework. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The goal of this research is to offer an effective intelligent model for forecasting college students’ career decisions in order to give a useful reference for career decisions and policy formation by relevant departments. The suggested prediction model is mainly based on a support vector machine (SVM) that has been modified using an enhanced butterfly optimization approach with a communication mechanism and Gaussian bare-bones mechanism (CBBOA). To get a better set of parameters and feature subsets, first, we added a communication mechanism to BOA to improve its global search capability and balance exploration and exploitation trends. Then, Gaussian bare-bones was added to increase the population diversity of BOA and its ability to jump out of the local optimum. The optimal SVM model (CBBOA-SVM) was then developed to predict the career decisions of college students based on the obtained parameters and feature subsets that are already optimized by CBBOA. In order to verify the effectiveness of CBBOA, we compared it with some advanced algorithms on all benchmark functions of CEC2014. Simulation results demonstrated that the performance of CBBOA is indeed more comprehensive. Meanwhile, comparisons between CBBOA-SVM and other machine learning approaches for career decision prediction were carried out, and the findings demonstrate that the provided CBBOA-SVM has better classification and more stable performance. As a result, it is plausible to conclude that the CBBOA-SVM is capable of being an effective tool for predicting college student career decisions.
Collapse
|
62
|
Zhang C, Mou M, Zhou Y, Zhang W, Lian X, Shi S, Lu M, Sun H, Li F, Wang Y, Zeng Z, Li Z, Zhang B, Qiu Y, Zhu F, Gao J. Biological activities of drug inactive ingredients. Brief Bioinform 2022; 23:6582006. [PMID: 35524477 DOI: 10.1093/bib/bbac160] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
In a drug formulation (DFM), the major components by mass are not Active Pharmaceutical Ingredient (API) but rather Drug Inactive Ingredients (DIGs). DIGs can reach much higher concentrations than that achieved by API, which raises great concerns about their clinical toxicities. Therefore, the biological activities of DIG on physiologically relevant target are widely demanded by both clinical investigation and pharmaceutical industry. However, such activity data are not available in any existing pharmaceutical knowledge base, and their potentials in predicting the DIG-target interaction have not been evaluated yet. In this study, the comprehensive assessment and analysis on the biological activities of DIGs were therefore conducted. First, the largest number of DIGs and DFMs were systematically curated and confirmed based on all drugs approved by US Food and Drug Administration. Second, comprehensive activities for both DIGs and DFMs were provided for the first time to pharmaceutical community. Third, the biological targets of each DIG and formulation were fully referenced to available databases that described their pharmaceutical/biological characteristics. Finally, a variety of popular artificial intelligence techniques were used to assess the predictive potential of DIGs' activity data, which was the first evaluation on the possibility to predict DIG's activity. As the activities of DIGs are critical for current pharmaceutical studies, this work is expected to have significant implications for the future practice of drug discovery and precision medicine.
Collapse
Affiliation(s)
- Chenyang Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China.,State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, China
| | - Wei Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xichen Lian
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shuiyang Shi
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Mingkun Lu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhenyu Zeng
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Zhaorong Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Bing Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| |
Collapse
|
63
|
Rani P, Dutta K, Kumar V. Artificial intelligence techniques for prediction of drug synergy in malignant diseases: Past, present, and future. Comput Biol Med 2022; 144:105334. [DOI: 10.1016/j.compbiomed.2022.105334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/13/2022] [Accepted: 02/13/2022] [Indexed: 12/22/2022]
|
64
|
An efficient rotational direction heap-based optimization with orthogonal structure for medical diagnosis. Comput Biol Med 2022; 146:105563. [PMID: 35551010 DOI: 10.1016/j.compbiomed.2022.105563] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 12/17/2022]
Abstract
The heap-based optimizer (HBO) is an optimization method proposed in recent years that may face local stagnation problems and show slow convergence speed due to the lack of detailed analysis of optimal solutions and a comprehensive search. Therefore, to mitigate these drawbacks and strengthen the performance of the algorithm in the field of medical diagnosis, a new MGOHBO method is proposed by introducing the modified Rosenbrock's rotational direction method (MRM), an operator from the grey wolf optimizer (GWM), and an orthogonal learning strategy (OL). The MGOHBO is compared with eleven famous and improved optimizers on IEEE CEC 2017. The results on benchmark functions indicate that the boosted MGOHBO has several significant advantages in terms of convergence accuracy and speed of the process. Additionally, this article analyzed the diversity and balance of MGOHBO in detail. Finally, the proposed MGOHBO algorithm is utilized to optimize the kernel extreme learning machines (KELM), and a new MGOHBO-KELM is proposed. To validate the performance of MGOHBO-KELM, seven disease diagnostic questions were introduced for testing in this work. In contrast to advanced models such as HBO-KELM and BP, it can be concluded that the MGOHBO-KELM model can achieve optimal results, which also proves that it has practical significance in solving medical diagnosis problems.
Collapse
|
65
|
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: 1.5] [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
|
66
|
Yang X, Zhao D, Yu F, Heidari AA, Bano Y, Ibrohimov A, Liu Y, Cai Z, Chen H, Chen X. An optimized machine learning framework for predicting intradialytic hypotension using indexes of chronic kidney disease-mineral and bone disorders. Comput Biol Med 2022; 145:105510. [DOI: 10.1016/j.compbiomed.2022.105510] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 11/03/2022]
|
67
|
Chen Y, Wang Y, Ding Y, Su X, Wang C. RGCNCDA: Relational graph convolutional network improves circRNA-disease association prediction by incorporating microRNAs. Comput Biol Med 2022; 143:105322. [PMID: 35217342 DOI: 10.1016/j.compbiomed.2022.105322] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/21/2022]
Abstract
Recently, a large number of studies have indicated that circRNAs with covalently closed loops play important roles in biological processes and have potential as diagnostic biomarkers. Therefore, research on the circRNA-disease relationship is helpful in disease diagnosis and treatment. However, traditional biological verification methods require considerable labor and time costs. In this paper, we propose a new computational method (RGCNCDA) to predict circRNA-disease associations based on relational graph convolutional networks (R-GCNs). The method first integrates the circRNA similarity network, miRNA similarity network, disease similarity network and association networks among them to construct a global heterogeneous network. Then, it employs the random walk with restart (RWR) and principal component analysis (PCA) models to learn low-dimensional and high-order information from the global heterogeneous network as the topological features. Finally, a prediction model based on an R-GCN encoder and a DistMult decoder is built to predict the potential disease-associated circRNA. The predicted results demonstrate that RGCNCDA performs significantly better than the other six state-of-the-art methods in a 5-fold cross validation. Furthermore, the case study illustrates that RGCNCDA can effectively discover potential circRNA-disease associations.
Collapse
Affiliation(s)
- Yaojia Chen
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Yanpeng Wang
- Beidahuang Industry Group General Hospital, Harbin, China
| | - Yijie Ding
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Xi Su
- Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China.
| | - Chunyu Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
68
|
Random Replacement Crisscross Butterfly Optimization Algorithm for Standard Evaluation of Overseas Chinese Associations. ELECTRONICS 2022. [DOI: 10.3390/electronics11071080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The butterfly optimization algorithm (BOA) is a swarm intelligence optimization algorithm proposed in 2019 that simulates the foraging behavior of butterflies. Similarly, the BOA itself has certain shortcomings, such as a slow convergence speed and low solution accuracy. To cope with these problems, two strategies are introduced to improve the performance of BOA. One is the random replacement strategy, which involves replacing the position of the current solution with that of the optimal solution and is used to increase the convergence speed. The other is the crisscross search strategy, which is utilized to trade off the capability of exploration and exploitation in BOA to remove local dilemmas whenever possible. In this case, we propose a novel optimizer named the random replacement crisscross butterfly optimization algorithm (RCCBOA). In order to evaluate the performance of RCCBOA, comparative experiments are conducted with another nine advanced algorithms on the IEEE CEC2014 function test set. Furthermore, RCCBOA is combined with support vector machine (SVM) and feature selection (FS)—namely, RCCBOA-SVM-FS—to attain a standardized construction model of overseas Chinese associations. It is found that the reasonableness of bylaws; the regularity of general meetings; and the right to elect, be elected, and vote are of importance to the planning and standardization of Chinese associations. Compared with other machine learning methods, the RCCBOA-SVM-FS model has an up to 95% accuracy when dealing with the normative prediction problem of overseas Chinese associations. Therefore, the constructed model is helpful for guiding the orderly and healthy development of overseas Chinese associations.
Collapse
|
69
|
Xia W, Zheng L, Fang J, Li F, Zhou Y, Zeng Z, Zhang B, Li Z, Li H, Zhu F. PFmulDL: a novel strategy enabling multi-class and multi-label protein function annotation by integrating diverse deep learning methods. Comput Biol Med 2022; 145:105465. [PMID: 35366467 DOI: 10.1016/j.compbiomed.2022.105465] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Bioinformatic annotation of protein function is essential but extremely sophisticated, which asks for extensive efforts to develop effective prediction method. However, the existing methods tend to amplify the representativeness of the families with large number of proteins by misclassifying the proteins in the families with small number of proteins. That is to say, the ability of the existing methods to annotate proteins in the 'rare classes' remains limited. Herein, a new protein function annotation strategy, PFmulDL, integrating multiple deep learning methods, was thus constructed. First, the recurrent neural network was integrated, for the first time, with the convolutional neural network to facilitate the function annotation. Second, a transfer learning method was introduced to the model construction for further improving the prediction performances. Third, based on the latest data of Gene Ontology, the newly constructed model could annotate the largest number of protein families comparing with the existing methods. Finally, this newly constructed model was found capable of significantly elevating the prediction performance for the 'rare classes' without sacrificing that for the 'major classes'. All in all, due to the emerging requirements on improving the prediction performance for the proteins in 'rare classes', this new strategy would become an essential complement to the existing methods for protein function prediction. All the models and source codes are freely available and open to all users at: https://github.com/idrblab/PFmulDL.
Collapse
Affiliation(s)
- Weiqi Xia
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Lingyan Zheng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Jiebin Fang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Zhenyu Zeng
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Bing Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Zhaorong Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Honglin Li
- School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China.
| |
Collapse
|
70
|
HKAM-MKM: A hybrid kernel alignment maximization-based multiple kernel model for identifying DNA-binding proteins. Comput Biol Med 2022; 145:105395. [PMID: 35334314 DOI: 10.1016/j.compbiomed.2022.105395] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/24/2022]
Abstract
The identification of DNA-binding proteins (DBPs) has always been a hot issue in the field of sequence classification. However, considering that the experimental identification method is very resource-intensive, the construction of a computational prediction model is worthwhile. This study developed and evaluated a hybrid kernel alignment maximization-based multiple kernel model (HKAM-MKM) for predicting DBPs. First, we collected two datasets and performed feature extraction on the sequences to obtain six feature groups, and then constructed the corresponding kernels. To ensure the effective utilisation of the base kernel and avoid ignoring the difference between the sample and its neighbours, we proposed local kernel alignment to calculate the kernel between the sample and its neighbours, with each sample as the centre. We combined the global and local kernel alignments to develop a hybrid kernel alignment model, and balance the relationship between the two through parameters. By maximising the hybrid kernel alignment value, we obtained the weight of each kernel and then linearly combined the kernels in the form of weights. Finally, the fused kernel was input into a support vector machine for training and prediction. Finally, in the independent test sets PDB186 and PDB2272, we obtained the highest Matthew's correlation coefficient (MCC) (0.768 and 0.5962, respectively) and the highest accuracy (87.1% and 78.43%, respectively), which were superior to the other predictors. Therefore, HKAM-MKM is an efficient prediction tool for DBPs.
Collapse
|
71
|
Vardhan S, Sahoo SK. Computational studies on the interaction of SARS-CoV-2 Omicron SGp RBD with human receptor ACE2, limonin and glycyrrhizic acid. Comput Biol Med 2022; 144:105367. [PMID: 35247766 PMCID: PMC8886687 DOI: 10.1016/j.compbiomed.2022.105367] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 01/06/2023]
Abstract
On November 24, 2021, the SARS-CoV-2 Omicron variant (B.1.1.529) was first identified in South Africa. The World Health Organization (WHO) declared the Omicron as a variant of concern (VoC) because of the unexpected and large numbers of mutations occurred in the genome, higher viral transmission and immune evasions. The present study was performed to explore the interactions of SARS-CoV-2 spike glycoprotein receptor-binding domain (SGp RBD) of the three variants (Omicron, Delta, and WT) with the receptor hACE2. The structural changes occurred in Omicron due to the mutations at key positions improved the ability to mediate SARS-CoV-2 viral infection compared to other VoCs. The phytochemicals limonin and glycyrrhizic acid were docked with the SGp RBD of the variants WT, Delta and Omicron. The computed dock score revealed that limonin and glycyrrhizic acid binds effectively at the SGp RBD of all three variants, and showed almost similar binding affinity at the binding interface of ACE2. Therefore, despite the multiple mutations occurred in Omicron and its viral transmission is comparatively high, the computed binding affinity of the phytochemicals limonin and glycyrrhizic acid supported that the traditional medicines can be useful in formulating adjuvant therapies to fight against the SARS-CoV-2 Omicron.
Collapse
Affiliation(s)
- Seshu Vardhan
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology (SVNIT), Surat, 395007, Gujarat, India
| | - Suban K Sahoo
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology (SVNIT), Surat, 395007, Gujarat, India.
| |
Collapse
|
72
|
Li F, Zhou Y, Zhang Y, Yin J, Qiu Y, Gao J, Zhu F. POSREG: proteomic signature discovered by simultaneously optimizing its reproducibility and generalizability. Brief Bioinform 2022; 23:6532538. [PMID: 35183059 DOI: 10.1093/bib/bbac040] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022] Open
Abstract
Mass spectrometry-based proteomic technique has become indispensable in current exploration of complex and dynamic biological processes. Instrument development has largely ensured the effective production of proteomic data, which necessitates commensurate advances in statistical framework to discover the optimal proteomic signature. Current framework mainly emphasizes the generalizability of the identified signature in predicting the independent data but neglects the reproducibility among signatures identified from independently repeated trials on different sub-dataset. These problems seriously restricted the wide application of the proteomic technique in molecular biology and other related directions. Thus, it is crucial to enable the generalizable and reproducible discovery of the proteomic signature with the subsequent indication of phenotype association. However, no such tool has been developed and available yet. Herein, an online tool, POSREG, was therefore constructed to identify the optimal signature for a set of proteomic data. It works by (i) identifying the proteomic signature of good reproducibility and aggregating them to ensemble feature ranking by ensemble learning, (ii) assessing the generalizability of ensemble feature ranking to acquire the optimal signature and (iii) indicating the phenotype association of discovered signature. POSREG is unique in its capacity of discovering the proteomic signature by simultaneously optimizing its reproducibility and generalizability. It is now accessible free of charge without any registration or login requirement at https://idrblab.org/posreg/.
Collapse
Affiliation(s)
- Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Ying Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Jianqing Gao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
73
|
Abstract
The dragonfly algorithm is a swarm intelligence optimization algorithm based on simulating the swarming behavior of dragonfly individuals. An efficient algorithm must have a symmetry of information between the participating entities. An improved dragonfly algorithm is proposed in this paper to further improve the global searching ability and the convergence speed of DA. The improved DA is named GGBDA, which adds Gaussian mutation and Gaussian barebone on the basis of DA. Gaussian mutation can randomly update the individual positions to avoid the algorithm falling into a local optimal solution. Gaussian barebone can quicken the convergent speed and strengthen local exploitation capacities. Enhancing algorithm efficiency relative to the symmetric concept is a critical challenge in the field of engineering design. To verify the superiorities of GGBDA, this paper sets 30 benchmark functions, which are taken from CEC2014 and 4 engineering design problems to compare GGBDA with other algorithms. The experimental result show that the Gaussian mutation and Gaussian barebone can effectively improve the performance of DA. The proposed GGBDA, similar to the DA, presents improvements in global optimization competence, search accuracy, and convergence performance.
Collapse
|
74
|
Zhang S, Amahong K, Zhang C, Li F, Gao J, Qiu Y, Zhu F. RNA-RNA interactions between SARS-CoV-2 and host benefit viral development and evolution during COVID-19 infection. Brief Bioinform 2022; 23:bbab397. [PMID: 34585235 PMCID: PMC8500159 DOI: 10.1093/bib/bbab397] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/11/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Some studies reported that genomic RNA of SARS-CoV-2 can absorb a few host miRNAs that regulate immune-related genes and then deprive their function. In this perspective, we conjecture that the absorption of the SARS-CoV-2 genome to host miRNAs is not a coincidence, which may be an indispensable approach leading to viral survival and development in host. In our study, we collected five datasets of miRNAs that were predicted to interact with the genome of SARS-CoV-2. The targets of these miRNAs in the five groups were consistently enriched immune-related pathways and virus-infectious diseases. Interestingly, the five datasets shared no one miRNA but their targets shared 168 genes. The signaling pathway enrichment of 168 shared targets implied an unbalanced immune response that the most of interleukin signaling pathways and none of the interferon signaling pathways were significantly different. Protein-protein interaction (PPI) network using the shared targets showed that PPI pairs, including IL6-IL6R, were related to the process of SARS-CoV-2 infection and pathogenesis. In addition, we found that SARS-CoV-2 absorption to host miRNA could benefit two popular mutant strains for more infectivity and pathogenicity. Conclusively, our results suggest that genomic RNA absorption to host miRNAs may be a vital approach by which SARS-CoV-2 disturbs the host immune system and infects host cells.
Collapse
Affiliation(s)
- Song Zhang
- College of Pharmaceutical Sciences in Zhejiang University, and the First Affiliated Hospital of Zhejiang University School of Medicine, China
| | | | - Chenyang Zhang
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Yunqing Qiu
- First Affiliated Hospital in Zhejiang University, China
| | - Feng Zhu
- College of Pharmaceutical Sciences in Zhejiang University, China
| |
Collapse
|
75
|
Wei L, Long W, Wei L. MDL-CPI: multi-view deep learning model for compound-protein interaction prediction. Methods 2022; 204:418-427. [PMID: 35114401 DOI: 10.1016/j.ymeth.2022.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022] Open
Abstract
Elucidating the mechanisms of Compound-Protein Interactions (CPIs) plays an essential role in drug discovery and development. Many computational efforts have been done to accelerate the development of this field. However, the current predictive performance is still not satisfactory, and existing methods consider only protein and compound features, ignoring their interactive information. In this study, we propose a multi-view deep learning method named MDL-CPI for CPI prediction. To sufficiently extract discriminative information, we introduce a hybrid architecture that leverages BERT (Bidirectional Encoder Representations from Transformers) and CNN (Convolutional Neural Network) to extract protein features from a sequential perspective, use the GNN (Graph Neural Networks) to extract compound features from a structural perspective, and generate a unified feature space by using AE2 (Autoencoder in Autoencoder Networks) network to learn the interactive information between BERT-CNN and Graph embeddings. Comparative results on benchmark datasets show that our proposed method exhibits better performance compared to existing CPI prediction methods, demonstrating the strong predictive ability of our model. Importantly, we demonstrate that the learned interactive information between compounds and proteins is critical to improve predictive performance. We release our source code and dataset at: https://github.com/Longwt123/MDL-CPI.
Collapse
|
76
|
Lin X. Genomic Variation Prediction: A Summary From Different Views. Front Cell Dev Biol 2021; 9:795883. [PMID: 34901036 PMCID: PMC8656232 DOI: 10.3389/fcell.2021.795883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/11/2021] [Indexed: 12/02/2022] Open
Abstract
Structural variations in the genome are closely related to human health and the occurrence and development of various diseases. To understand the mechanisms of diseases, find pathogenic targets, and carry out personalized precision medicine, it is critical to detect such variations. The rapid development of high-throughput sequencing technologies has accelerated the accumulation of large amounts of genomic mutation data, including synonymous mutations. Identifying pathogenic synonymous mutations that play important roles in the occurrence and development of diseases from all the available mutation data is of great importance. In this paper, machine learning theories and methods are reviewed, efficient and accurate pathogenic synonymous mutation prediction methods are developed, and a standardized three-level variant analysis framework is constructed. In addition, multiple variation tolerance prediction models are studied and integrated, and new ideas for structural variation detection based on deep information mining are explored.
Collapse
Affiliation(s)
- Xiuchun Lin
- College of Information and Electrical Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
77
|
Fu T, Li F, Zhang Y, Yin J, Qiu W, Li X, Liu X, Xin W, Wang C, Yu L, Gao J, Zheng Q, Zeng S, Zhu F. VARIDT 2.0: structural variability of drug transporter. Nucleic Acids Res 2021; 50:D1417-D1431. [PMID: 34747471 PMCID: PMC8728241 DOI: 10.1093/nar/gkab1013] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/08/2021] [Accepted: 11/04/2021] [Indexed: 12/20/2022] Open
Abstract
The structural variability data of drug transporter (DT) are key for research on precision medicine and rational drug use. However, these valuable data are not sufficiently covered by the available databases. In this study, a major update of VARIDT (a database previously constructed to provide DTs' variability data) was thus described. First, the experimentally resolved structures of all DTs reported in the original VARIDT were discovered from PubMed and Protein Data Bank. Second, the structural variability data of each DT were collected by literature review, which included: (a) mutation-induced spatial variations in folded state, (b) difference among DT structures of human and model organisms, (c) outward/inward-facing DT conformations and (d) xenobiotics-driven alterations in the 3D complexes. Third, for those DTs without experimentally resolved structural variabilities, homology modeling was further applied as well-established protocol to enrich such valuable data. As a result, 145 mutation-induced spatial variations of 42 DTs, 1622 inter-species structures originating from 292 DTs, 118 outward/inward-facing conformations belonging to 59 DTs, and 822 xenobiotics-regulated structures in complex with 57 DTs were updated to VARIDT (https://idrblab.org/varidt/ and http://varidt.idrblab.net/). All in all, the newly collected structural variabilities will be indispensable for explaining drug sensitivity/selectivity, bridging preclinical research with clinical trial, revealing the mechanism underlying drug-drug interaction, and so on.
Collapse
Affiliation(s)
- Tingting Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenqi Qiu
- Department of Surgery, HKU-SZH & Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xuedong Li
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xingang Liu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Wenwen Xin
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Chengzhao Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Qingchuan Zheng
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
78
|
Zhou Y, Zhang Y, Lian X, Li F, Wang C, Zhu F, Qiu Y, Chen Y. Therapeutic target database update 2022: facilitating drug discovery with enriched comparative data of targeted agents. Nucleic Acids Res 2021; 50:D1398-D1407. [PMID: 34718717 PMCID: PMC8728281 DOI: 10.1093/nar/gkab953] [Citation(s) in RCA: 319] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 11/14/2022] Open
Abstract
Drug discovery relies on the knowledge of not only drugs and targets, but also the comparative agents and targets. These include poor binders and non-binders for developing discovery tools, prodrugs for improved therapeutics, co-targets of therapeutic targets for multi-target strategies and off-target investigations, and the collective structure-activity and drug-likeness landscapes of enhanced drug feature. However, such valuable data are inadequately covered by the available databases. In this study, a major update of the Therapeutic Target Database, previously featured in NAR, was therefore introduced. This update includes (a) 34 861 poor binders and 12 683 non-binders of 1308 targets; (b) 534 prodrug-drug pairs for 121 targets; (c) 1127 co-targets of 672 targets regulated by 642 approved and 624 clinical trial drugs; (d) the collective structure-activity landscapes of 427 262 active agents of 1565 targets; (e) the profiles of drug-like properties of 33 598 agents of 1102 targets. Moreover, a variety of additional data and function are provided, which include the cross-links to the target structure in PDB and AlphaFold, 159 and 1658 newly emerged targets and drugs, and the advanced search function for multi-entry target sequences or drug structures. The database is accessible without login requirement at: https://idrblab.org/ttd/.
Collapse
Affiliation(s)
- Ying Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, China
| | - Yintao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xichen Lian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chaoxin Wang
- Department of Computer Science, Kansas State University, Manhattan 66506, USA
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, China
| | - Yuzong Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.,Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| |
Collapse
|
79
|
Subtype-selective mechanisms of negative allosteric modulators binding to group I metabotropic glutamate receptors. Acta Pharmacol Sin 2021; 42:1354-1367. [PMID: 33122823 PMCID: PMC8285414 DOI: 10.1038/s41401-020-00541-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Group I metabotropic glutamate receptors (mGlu1 and mGlu5) are promising targets for multiple psychiatric and neurodegenerative disorders. Understanding the subtype selectivity of mGlu1 and mGlu5 allosteric sites is essential for the rational design of novel modulators with single- or dual-target mechanism of action. In this study, starting from the deposited mGlu1 and mGlu5 crystal structures, we utilized computational modeling approaches integrating docking, molecular dynamics simulation, and efficient post-trajectory analysis to reveal the subtype-selective mechanism of mGlu1 and mGlu5 to 10 diverse drug scaffolds representing known negative allosteric modulators (NAMs) in the literature. The results of modeling identified six pairs of non-conserved residues and four pairs of conserved ones as critical features to distinguish the selective NAMs binding to the corresponding receptors. In addition, nine pairs of residues are beneficial to the development of novel dual-target NAMs of group I metabotropic glutamate receptors. Furthermore, the binding modes of a reported dual-target NAM (VU0467558) in mGlu1 and mGlu5 were predicted to verify the identified residues that play key roles in the receptor selectivity and the dual-target binding. The results of this study can guide rational structure-based design of novel NAMs, and the approach can be generally applicable to characterize the features of selectivity for other G-protein-coupled receptors.
Collapse
|
80
|
CWLy-RF: A novel approach for identifying cell wall lyases based on random forest classifier. Genomics 2021; 113:2919-2924. [PMID: 34186189 DOI: 10.1016/j.ygeno.2021.06.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/20/2021] [Accepted: 06/25/2021] [Indexed: 02/05/2023]
Abstract
Drug resistance of pathogenic bacteria has become increasingly serious due to the abuse of antibiotics in recent years. Researchers have found that cell wall lyases are effective antibacterial agents that can specifically recognize target bacteria and degrade bacterial peptidoglycan. Traditional wet experiments are usually expensive, time-consuming and laborious for the identification of lyases. Therefore, there is an urgent need to develop prediction tools based on computer methods to identify lyases quickly and accurately. In this paper, a new predictor, CWLy-RF, is proposed based on the random forest (RF) algorithm to identify cell wall lyases. In this method, we combined three features, namely, 400D, 188D and the composition of k-spaced amino acid group pairs, using mixed-feature representation methods. Afterward, we improved the feature representation ability with the selected top 100 features by using the information gain method and trained a predictive model using RF. The constructed prediction model is evaluated by using 10-fold cross-validation. The accuracy obtained was 96.09%, the AUC was 0.993, the MCC was 0.922, the sensitivity was 94.92%, and the specificity was 97.32%. We have proved that the proposed predictor CWLy-RF is superior to other latest models, and it will hopefully become an effective and useful tool for identifying lyases.
Collapse
|
81
|
Vallespi MG, Mestre B, Marrero MA, Uranga R, Rey D, Lugiollo M, Betancourt M, Silva K, Corrales D, Lamadrid Y, Rodriguez Y, Maceo A, Chaviano PP, Lemos G, Cabrales A, Freyre FM, Santana H, Garay HE, Oliva B, Fernandez JR. A first-in-class, first-in-human, phase I trial of CIGB-552, a synthetic peptide targeting COMMD1 to inhibit the oncogenic activity of NF-κB in patients with advanced solid tumors. Int J Cancer 2021; 149:1313-1321. [PMID: 34019700 DOI: 10.1002/ijc.33695] [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: 02/09/2021] [Revised: 04/05/2021] [Accepted: 04/29/2021] [Indexed: 11/06/2022]
Abstract
CIGB-552 is a synthetic peptide that interacts with COMMD1 and upregulates its protein levels. The objectives of this phase I study were safety, pharmacokinetic profile, evaluation of the lymphocytes CD4+ and CD8+ and preliminary activity in patients with advanced tumors. A 3 + 3 dose-escalation design with seven dose levels was implemented. Patients were included until a grade 3 related adverse event occurred and the maximum tolerated dose was reached. The patients received subcutaneous administration of CIGB-552 three times per week for 2 weeks. Single-dose plasma pharmacokinetics was characterized at two dose levels, and tumor responses were classified by RECIST 1.1. Twenty-four patients received CIGB-552. Dose-limiting toxicity was associated with a transient grade 3 pruritic maculopapular rash at a dose of 7.0 mg. The maximum tolerated dose was defined as 4.7 mg. Ten patients were assessable for immunological status. Seven patients had significant changes in the ratio CD4/CD8 in response to CIGB-552 treatment; three patients did not modify the immunological status. Stable disease was observed in five patients, including two metastatic soft sarcomas. We conclude that CIGB-552 at dose 4.7 mg was well tolerated with no significant adverse events and appeared to provide some clinical benefits.
Collapse
Affiliation(s)
| | - Braulio Mestre
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Maria A Marrero
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Rolando Uranga
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Diana Rey
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Martha Lugiollo
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Mircea Betancourt
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Kirenia Silva
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Danay Corrales
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Yanet Lamadrid
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Yamilka Rodriguez
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Anaelys Maceo
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Pedro P Chaviano
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Gilda Lemos
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Ania Cabrales
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Freya M Freyre
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Hector Santana
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Hilda E Garay
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Brizaida Oliva
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Julio R Fernandez
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| |
Collapse
|
82
|
Yin J, Li X, Li F, Lu Y, Zeng S, Zhu F. Identification of the key target profiles underlying the drugs of narrow therapeutic index for treating cancer and cardiovascular disease. Comput Struct Biotechnol J 2021; 19:2318-2328. [PMID: 33995923 PMCID: PMC8105181 DOI: 10.1016/j.csbj.2021.04.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 12/14/2022] Open
Abstract
An appropriate therapeutic index is crucial for drug discovery and development since narrow therapeutic index (NTI) drugs with slight dosage variation may induce severe adverse drug reactions or potential treatment failure. To date, the shared characteristics underlying the targets of NTI drugs have been explored by several studies, which have been applied to identify potential drug targets. However, the association between the drug therapeutic index and the related disease has not been dissected, which is important for revealing the NTI drug mechanism and optimizing drug design. Therefore, in this study, two classes of disease (cancers and cardiovascular disorders) with the largest number of NTI drugs were selected, and the target property of the corresponding NTI drugs was analyzed. By calculating the biological system profiles and human protein–protein interaction (PPI) network properties of drug targets and adopting an AI-based algorithm, differentiated features between two diseases were discovered to reveal the distinct underlying mechanisms of NTI drugs in different diseases. Consequently, ten shared features and four unique features were identified for both diseases to distinguish NTI from NNTI drug targets. These computational discoveries, as well as the newly found features, suggest that in the clinical study of avoiding narrow therapeutic index in those diseases, the ability of target to be a hub and the efficiency of target signaling in the human PPI network should be considered, and it could thus provide novel guidance in the drug discovery and clinical research process and help to estimate the drug safety of cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoxu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China.,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
83
|
Fu J, Zhang Y, Liu J, Lian X, Tang J, Zhu F. Pharmacometabonomics: data processing and statistical analysis. Brief Bioinform 2021; 22:6236068. [PMID: 33866355 DOI: 10.1093/bib/bbab138] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/09/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Individual variations in drug efficacy, side effects and adverse drug reactions are still challenging that cannot be ignored in drug research and development. The aim of pharmacometabonomics is to better understand the pharmacokinetic properties of drugs and monitor the drug effects on specific metabolic pathways. Here, we systematically reviewed the recent technological advances in pharmacometabonomics for better understanding the pathophysiological mechanisms of diseases as well as the metabolic effects of drugs on bodies. First, the advantages and disadvantages of all mainstream analytical techniques were compared. Second, many data processing strategies including filtering, missing value imputation, quality control-based correction, transformation, normalization together with the methods implemented in each step were discussed. Third, various feature selection and feature extraction algorithms commonly applied in pharmacometabonomics were described. Finally, the databases that facilitate current pharmacometabonomics were collected and discussed. All in all, this review provided guidance for researchers engaged in pharmacometabonomics and metabolomics, and it would promote the wide application of metabolomics in drug research and personalized medicine.
Collapse
Affiliation(s)
- Jianbo Fu
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Ying Zhang
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Jin Liu
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Xichen Lian
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Jing Tang
- Department of Bioinformatics in Chongqing Medical University, China
| | - Feng Zhu
- College of Pharmaceutical Sciences in Zhejiang University, China
| |
Collapse
|
84
|
Xu L, Jiao S, Zhang D, Wu S, Zhang H, Gao B. Identification of long noncoding RNAs with machine learning methods: a review. Brief Funct Genomics 2021; 20:174-180. [PMID: 33758917 DOI: 10.1093/bfgp/elab017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are noncoding RNAs with a length greater than 200 nucleotides. Studies have shown that they play an important role in many life activities. Dozens of lncRNAs have been characterized to some extent, and they are reported to be related to the development of diseases in a variety of cells. However, the biological functions of most lncRNAs are currently still unclear. Therefore, accurately identifying and predicting lncRNAs would be helpful for research on their biological functions. Due to the disadvantages of high cost and high resource-intensiveness of experimental methods, scientists have developed numerous computational methods to identify and predict lncRNAs in recent years. In this paper, we systematically summarize the machine learning-based lncRNAs prediction tools from several perspectives, and discuss the challenges and prospects for the future work.
Collapse
Affiliation(s)
- Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic
| | - Shihu Jiao
- College of Chemistry, Sichuan University, Sichuan, China
| | - Dandan Zhang
- Departments of Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University
| | - Song Wu
- Preventive Treatment of Disease Centre of Qinhuangdao Hospital of Traditional Chinese Medicine
| | - Haihong Zhang
- First Affiliated Hospital of Harbin Medical University
| | - Bo Gao
- Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
85
|
Zhang S, Amahong K, Sun X, Lian X, Liu J, Sun H, Lou Y, Zhu F, Qiu Y. The miRNA: a small but powerful RNA for COVID-19. Brief Bioinform 2021; 22:1137-1149. [PMID: 33675361 PMCID: PMC7989616 DOI: 10.1093/bib/bbab062] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a severe and rapidly evolving epidemic. Now, although a few drugs and vaccines have been proved for its treatment and prevention, little systematic comments are made to explain its susceptibility to humans. A few scattered studies used bioinformatics methods to explore the role of microRNA (miRNA) in COVID-19 infection. Combining these timely reports and previous studies about virus and miRNA, we comb through the available clues and seemingly make the perspective reasonable that the COVID-19 cleverly exploits the interplay between the small miRNA and other biomolecules to avoid being effectively recognized and attacked from host immune protection as well to deactivate functional genes that are crucial for immune system. In detail, SARS-CoV-2 can be regarded as a sponge to adsorb host immune-related miRNA, which forces host fall into dysfunction status of immune system. Besides, SARS-CoV-2 encodes its own miRNAs, which can enter host cell and are not perceived by the host's immune system, subsequently targeting host function genes to cause illnesses. Therefore, this article presents a reasonable viewpoint that the miRNA-based interplays between the host and SARS-CoV-2 may be the primary cause that SARS-CoV-2 accesses and attacks the host cells.
Collapse
Affiliation(s)
- Song Zhang
- College of Pharmaceutical Sciences in Zhejiang University and the First Affiliated Hospital of Zhejiang University School of Medicine, China
| | | | - Xiuna Sun
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Xichen Lian
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Jin Liu
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Yan Lou
- Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, the First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Feng Zhu
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, the First Affiliated Hospital, Zhejiang University School of Medicine, China
| |
Collapse
|
86
|
Recent Advances in Predicting Protein S-Nitrosylation Sites. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5542224. [PMID: 33628788 PMCID: PMC7892234 DOI: 10.1155/2021/5542224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 01/09/2023]
Abstract
Protein S-nitrosylation (SNO) is a process of covalent modification of nitric oxide (NO) and its derivatives and cysteine residues. SNO plays an essential role in reversible posttranslational modifications of proteins. The accurate prediction of SNO sites is crucial in revealing a certain biological mechanism of NO regulation and related drug development. Identification of the sites of SNO in proteins is currently a very hot topic. In this review, we briefly summarize recent advances in computationally identifying SNO sites. The challenges and future perspectives for identifying SNO sites are also discussed. We anticipate that this review will provide insights into research on SNO site prediction.
Collapse
|
87
|
Fu J, Luo Y, Mou M, Zhang H, Tang J, Wang Y, Zhu F. Advances in Current Diabetes Proteomics: From the Perspectives of Label- free Quantification and Biomarker Selection. Curr Drug Targets 2021; 21:34-54. [PMID: 31433754 DOI: 10.2174/1389450120666190821160207] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Due to its prevalence and negative impacts on both the economy and society, the diabetes mellitus (DM) has emerged as a worldwide concern. In light of this, the label-free quantification (LFQ) proteomics and diabetic marker selection methods have been applied to elucidate the underlying mechanisms associated with insulin resistance, explore novel protein biomarkers, and discover innovative therapeutic protein targets. OBJECTIVE The purpose of this manuscript is to review and analyze the recent computational advances and development of label-free quantification and diabetic marker selection in diabetes proteomics. METHODS Web of Science database, PubMed database and Google Scholar were utilized for searching label-free quantification, computational advances, feature selection and diabetes proteomics. RESULTS In this study, we systematically review the computational advances of label-free quantification and diabetic marker selection methods which were applied to get the understanding of DM pathological mechanisms. Firstly, different popular quantification measurements and proteomic quantification software tools which have been applied to the diabetes studies are comprehensively discussed. Secondly, a number of popular manipulation methods including transformation, pretreatment (centering, scaling, and normalization), missing value imputation methods and a variety of popular feature selection techniques applied to diabetes proteomic data are overviewed with objective evaluation on their advantages and disadvantages. Finally, the guidelines for the efficient use of the computationbased LFQ technology and feature selection methods in diabetes proteomics are proposed. CONCLUSION In summary, this review provides guidelines for researchers who will engage in proteomics biomarker discovery and by properly applying these proteomic computational advances, more reliable therapeutic targets will be found in the field of diabetes mellitus.
Collapse
Affiliation(s)
- Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongning Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jing Tang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| |
Collapse
|
88
|
Liang G, Wu J, Xu L. A prognosis-related based method for miRNA selection on liver hepatocellular carcinoma prediction. Comput Biol Chem 2021; 91:107433. [PMID: 33540232 DOI: 10.1016/j.compbiolchem.2020.107433] [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: 09/24/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is considered as the sixth most common cancer in the world, and it is also considered as one of the causes of death. Moreover, the poor prognosis of recurrence of HCC after surgery and metastasis is also a big problem for human health. If the disease can be diagnosed earlier, the survival rate of the patients will be improved significantly. In the early stage of hepatocellular carcinoma, the expression of miRNAs is likely to become abnormal. In our work, the expression profile of miRNAs of human HCC in cancer tissue is compared with their adjacent tissue samples collected from tumor cancer genomic Atlas (TCGA) platform, then the genes with significant difference are selected by Limma test. Selected genes are referred to predict miRNAs related to the prognosis of HCC patients. Finally, miRNAs regulated by target genes are selected by our method, and the experimental results demonstrated that our method is more efficient than biology wet experimental method with lower cost.
Collapse
Affiliation(s)
- Guangmin Liang
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, 518000, China
| | - Jin Wu
- School of Management, Shenzhen Polytechnic, Shenzhen, 518000, China.
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, 518000, China.
| |
Collapse
|
89
|
Yin J, Li F, Zhou Y, Mou M, Lu Y, Chen K, Xue J, Luo Y, Fu J, He X, Gao J, Zeng S, Yu L, Zhu F. INTEDE: interactome of drug-metabolizing enzymes. Nucleic Acids Res 2021; 49:D1233-D1243. [PMID: 33045737 PMCID: PMC7779056 DOI: 10.1093/nar/gkaa755] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/19/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Drug-metabolizing enzymes (DMEs) are critical determinant of drug safety and efficacy, and the interactome of DMEs has attracted extensive attention. There are 3 major interaction types in an interactome: microbiome-DME interaction (MICBIO), xenobiotics-DME interaction (XEOTIC) and host protein-DME interaction (HOSPPI). The interaction data of each type are essential for drug metabolism, and the collective consideration of multiple types has implication for the future practice of precision medicine. However, no database was designed to systematically provide the data of all types of DME interactions. Here, a database of the Interactome of Drug-Metabolizing Enzymes (INTEDE) was therefore constructed to offer these interaction data. First, 1047 unique DMEs (448 host and 599 microbial) were confirmed, for the first time, using their metabolizing drugs. Second, for these newly confirmed DMEs, all types of their interactions (3359 MICBIOs between 225 microbial species and 185 DMEs; 47 778 XEOTICs between 4150 xenobiotics and 501 DMEs; 7849 HOSPPIs between 565 human proteins and 566 DMEs) were comprehensively collected and then provided, which enabled the crosstalk analysis among multiple types. Because of the huge amount of accumulated data, the INTEDE made it possible to generalize key features for revealing disease etiology and optimizing clinical treatment. INTEDE is freely accessible at: https://idrblab.org/intede/.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kangli Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jia Xue
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xu He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| |
Collapse
|
90
|
Genome-Wide Analysis of LysM-Containing Gene Family in Wheat: Structural and Phylogenetic Analysis during Development and Defense. Genes (Basel) 2020; 12:genes12010031. [PMID: 33383636 PMCID: PMC7823900 DOI: 10.3390/genes12010031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 11/17/2022] Open
Abstract
The lysin motif (LysM) family comprise a number of defense proteins that play important roles in plant immunity. The LysM family includes LysM-containing receptor-like proteins (LYP) and LysM-containing receptor-like kinase (LYK). LysM generally recognizes the chitin and peptidoglycan derived from bacteria and fungi. Approximately 4000 proteins with the lysin motif (Pfam PF01476) are found in prokaryotes and eukaryotes. Our study identified 57 LysM genes and 60 LysM proteins in wheat and renamed these genes and proteins based on chromosome distribution. According to the phylogenetic and gene structure of intron-exon distribution analysis, the 60 LysM proteins were classified into seven groups. Gene duplication events had occurred among the LysM family members during the evolution process, resulting in an increase in the LysM gene family. Synteny analysis suggested the characteristics of evolution of the LysM family in wheat and other species. Systematic analysis of these species provided a foundation of LysM genes in crop defense. A comprehensive analysis of the expression and cis-elements of LysM gene family members suggested that they play an essential role in defending against plant pathogens. The present study provides an overview of the LysM family in the wheat genome as well as information on systematic, phylogenetic, gene duplication, and intron-exon distribution analyses that will be helpful for future functional analysis of this important protein family, especially in Gramineae species.
Collapse
|
91
|
Yang Q, Hong J, Li Y, Xue W, Li S, Yang H, Zhu F. A novel bioinformatics approach to identify the consistently well-performing normalization strategy for current metabolomic studies. Brief Bioinform 2020; 21:2142-2152. [PMID: 31776543 PMCID: PMC7711263 DOI: 10.1093/bib/bbz137] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/26/2019] [Accepted: 10/05/2019] [Indexed: 12/19/2022] Open
Abstract
Unwanted experimental/biological variation and technical error are frequently encountered in current metabolomics, which requires the employment of normalization methods for removing undesired data fluctuations. To ensure the 'thorough' removal of unwanted variations, the collective consideration of multiple criteria ('intragroup variation', 'marker stability' and 'classification capability') was essential. However, due to the limited number of available normalization methods, it is extremely challenging to discover the appropriate one that can meet all these criteria. Herein, a novel approach was proposed to discover the normalization strategies that are consistently well performing (CWP) under all criteria. Based on various benchmarks, all normalization methods popular in current metabolomics were 'first' discovered to be non-CWP. 'Then', 21 new strategies that combined the 'sample'-based method with the 'metabolite'-based one were found to be CWP. 'Finally', a variety of currently available methods (such as cubic splines, range scaling, level scaling, EigenMS, cyclic loess and mean) were identified to be CWP when combining with other normalization. In conclusion, this study not only discovered several strategies that performed consistently well under all criteria, but also proposed a novel approach that could ensure the identification of CWP strategies for future biological problems.
Collapse
Affiliation(s)
- Qingxia Yang
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| | - Jiajun Hong
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| | - Yi Li
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| | - Weiwei Xue
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| | - Song Li
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| | - Hui Yang
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| | - Feng Zhu
- Ph.D. candidates of Zhejiang University, China, and jointly cultivated by the School of Pharmaceutical Sciences in Chongqing University, China. Their main research interests include OMICs-based bioinformatics and statistical metabolomics
| |
Collapse
|
92
|
Hou R, Wu J, Xu L, Zou Q, Wu YJ. Computational Prediction of Protein Arginine Methylation Based on Composition-Transition-Distribution Features. ACS OMEGA 2020; 5:27470-27479. [PMID: 33134710 PMCID: PMC7594152 DOI: 10.1021/acsomega.0c03972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Arginine methylation is one of the most essential protein post-translational modifications. Identifying the site of arginine methylation is a critical problem in biology research. Unfortunately, biological experiments such as mass spectrometry are expensive and time-consuming. Hence, predicting arginine methylation by machine learning is an alternative fast and efficient way. In this paper, we focus on the systematic characterization of arginine methylation with composition-transition-distribution (CTD) features. The presented framework consists of three stages. In the first stage, we extract CTD features from 1750 samples and exploit decision tree to generate accurate prediction. The accuracy of prediction can reach 96%. In the second stage, the support vector machine can predict the number of arginine methylation sites with 0.36 R-squared. In the third stage, experiments carried out with the updated arginine methylation site data set show that utilizing CTD features and adopting random forest as the classifier outperform previous methods. The accuracy of identification can reach 82.1 and 82.5% in single methylarginine and double methylarginine data sets, respectively. The discovery presented in this paper can be helpful for future research on arginine methylation.
Collapse
Affiliation(s)
- Ruiyan Hou
- Laboratory
of Molecular Toxicology, State Key Laboratory of Integrated Management
of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Science, University of Chinese Academy
of Sciences, Beijing 100049, China
| | - Jin Wu
- School
of Management, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Lei Xu
- School
of Electronic and Engineering, Shenzhen
Polytechnic, Shenzhen 518055, China
| | - Quan Zou
- Institute
of Fundamental and Frontier Sciences, University
of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yi-Jun Wu
- Laboratory
of Molecular Toxicology, State Key Laboratory of Integrated Management
of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
93
|
Guo Z, Wang P, Liu Z, Zhao Y. Discrimination of Thermophilic Proteins and Non-thermophilic Proteins Using Feature Dimension Reduction. Front Bioeng Biotechnol 2020; 8:584807. [PMID: 33195148 PMCID: PMC7642589 DOI: 10.3389/fbioe.2020.584807] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/11/2020] [Indexed: 01/19/2023] Open
Abstract
Thermophilicity is a very important property of proteins, as it sometimes determines denaturation and cell death. Thus, methods for predicting thermophilic proteins and non-thermophilic proteins are of interest and can contribute to the design and engineering of proteins. In this article, we describe the use of feature dimension reduction technology and LIBSVM to identify thermophilic proteins. The highest accuracy obtained by cross-validation was 96.02% with 119 parameters. When using only 16 features, we obtained an accuracy of 93.33%. We discuss the importance of the different characteristics in identification and report a comparison of the performance of support vector machine to that of other methods.
Collapse
Affiliation(s)
- Zifan Guo
- School of Aeronautics and Astronautic, Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Pingping Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhendong Liu
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Yuming Zhao
- Information and Computer Engineering College, Northeast Forestry University, Harbin, China
| |
Collapse
|
94
|
A Method for Identifying Vesicle Transport Proteins Based on LibSVM and MRMD. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:8926750. [PMID: 33133228 PMCID: PMC7591939 DOI: 10.1155/2020/8926750] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/14/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
With the development of computer technology, many machine learning algorithms have been applied to the field of biology, forming the discipline of bioinformatics. Protein function prediction is a classic research topic in this subject area. Though many scholars have made achievements in identifying protein by different algorithms, they often extract a large number of feature types and use very complex classification methods to obtain little improvement in the classification effect, and this process is very time-consuming. In this research, we attempt to utilize as few features as possible to classify vesicular transportation proteins and to simultaneously obtain a comparative satisfactory classification result. We adopt CTDC which is a submethod of the method of composition, transition, and distribution (CTD) to extract only 39 features from each sequence, and LibSVM is used as the classification method. We use the SMOTE method to deal with the problem of dataset imbalance. There are 11619 protein sequences in our dataset. We selected 4428 sequences to train our classification model and selected other 1832 sequences from our dataset to test the classification effect and finally achieved an accuracy of 71.77%. After dimension reduction by MRMD, the accuracy is 72.16%.
Collapse
|
95
|
Xu L, Liang G, Chen B, Tan X, Xiang H, Liao C. A Computational Method for the Identification of Endolysins and Autolysins. Protein Pept Lett 2020; 27:329-336. [PMID: 31577192 DOI: 10.2174/0929866526666191002104735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/27/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cell lytic enzyme is a kind of highly evolved protein, which can destroy the cell structure and kill the bacteria. Compared with antibiotics, cell lytic enzyme will not cause serious problem of drug resistance of pathogenic bacteria. Thus, the study of cell wall lytic enzymes aims at finding an efficient way for curing bacteria infectious. Compared with using antibiotics, the problem of drug resistance becomes more serious. Therefore, it is a good choice for curing bacterial infections by using cell lytic enzymes. Cell lytic enzyme includes endolysin and autolysin and the difference between them is the purpose of the break of cell wall. The identification of the type of cell lytic enzymes is meaningful for the study of cell wall enzymes. OBJECTIVE In this article, our motivation is to predict the type of cell lytic enzyme. Cell lytic enzyme is helpful for killing bacteria, so it is meaningful for study the type of cell lytic enzyme. However, it is time consuming to detect the type of cell lytic enzyme by experimental methods. Thus, an efficient computational method for the type of cell lytic enzyme prediction is proposed in our work. METHODS We propose a computational method for the prediction of endolysin and autolysin. First, a data set containing 27 endolysins and 41 autolysins is built. Then the protein is represented by tripeptides composition. The features are selected with larger confidence degree. At last, the classifier is trained by the labeled vectors based on support vector machine. The learned classifier is used to predict the type of cell lytic enzyme. RESULTS Following the proposed method, the experimental results show that the overall accuracy can attain 97.06%, when 44 features are selected. Compared with Ding's method, our method improves the overall accuracy by nearly 4.5% ((97.06-92.9)/92.9%). The performance of our proposed method is stable, when the selected feature number is from 40 to 70. The overall accuracy of tripeptides optimal feature set is 94.12%, and the overall accuracy of Chou's amphiphilic PseAAC method is 76.2%. The experimental results also demonstrate that the overall accuracy is improved by nearly 18% when using the tripeptides optimal feature set. CONCLUSION The paper proposed an efficient method for identifying endolysin and autolysin. In this paper, support vector machine is used to predict the type of cell lytic enzyme. The experimental results show that the overall accuracy of the proposed method is 94.12%, which is better than some existing methods. In conclusion, the selected 44 features can improve the overall accuracy for identification of the type of cell lytic enzyme. Support vector machine performs better than other classifiers when using the selected feature set on the benchmark data set.
Collapse
Affiliation(s)
- Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Guangmin Liang
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Baowen Chen
- School of Software, Shenzhen Institute of Information Technology, Shenzhen, China
| | - Xu Tan
- School of Software, Shenzhen Institute of Information Technology, Shenzhen, China
| | - Huaikun Xiang
- School of Automotive and Transportation Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Changrui Liao
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
96
|
Li Q, Zhou W, Wang D, Wang S, Li Q. Prediction of Anticancer Peptides Using a Low-Dimensional Feature Model. Front Bioeng Biotechnol 2020; 8:892. [PMID: 32903381 PMCID: PMC7434836 DOI: 10.3389/fbioe.2020.00892] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/10/2020] [Indexed: 01/09/2023] Open
Abstract
Cancer is still a severe health problem globally. The therapy of cancer traditionally involves the use of radiotherapy or anticancer drugs to kill cancer cells, but these methods are quite expensive and have side effects, which will cause great harm to patients. With the find of anticancer peptides (ACPs), significant progress has been achieved in the therapy of tumors. Therefore, it is invaluable to accurately identify anticancer peptides. Although biochemical experiments can solve this work, this method is expensive and time-consuming. To promote the application of anticancer peptides in cancer therapy, machine learning can be used to recognize anticancer peptides by extracting the feature vectors of anticancer peptides. Nevertheless, poor performance usually be found in training the machine learning model to utilizing high-dimensional features in practice. In order to solve the above job, this paper put forward a 19-dimensional feature model based on anticancer peptide sequences, which has lower dimensionality and better performance than some existing methods. In addition, this paper also separated a model with a low number of dimensions and acceptable performance. The few features identified in this study may represent the important features of anticancer peptides.
Collapse
Affiliation(s)
- Qingwen Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Wenyang Zhou
- Center for Bioinformatics, School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, China
| | - Donghua Wang
- Department of General Surgery, Heilongjiang Province Land Reclamation Headquarters General Hospital, Harbin, China
| | - Sui Wang
- Key Laboratory of Soybean Biology in Chinese Ministry of Education, Northeast Agricultural University, Harbin, China
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
| | - Qingyuan Li
- Forestry and Fruit Tree Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
97
|
Wu C, Ono S. Exploratory Analysis of the Factors Associated With Success Rates of Confirmatory Randomized Controlled Trials in Cancer Drug Development. Clin Transl Sci 2020; 14:260-267. [PMID: 32702190 PMCID: PMC7877835 DOI: 10.1111/cts.12852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
This study examined the outcomes of recent confirmatory randomized controlled trials (RCTs) in phase III that were initiated between 2005 and 2017 for oncologic drugs in the United States and identified several factors that were associated with the success of RCTs. Our regression analysis showed that studies with progression‐free survival or response rate as primary end point were more likely to succeed than studies with overall survival (odds ratio (OR) = 2.94 and 6.23, respectively). The status of development was also linked with success rates. Studies for non‐lead indication tended to have lower success rates than studies for lead indication (OR = 0.68). Studies for first‐line therapy were observed to have low success rates compared with studies for post second‐line therapies (OR = 0.37). Studies for which strong prior evidence was not listed in their publication tended to be more successful than studies that followed rigorous RCTs or single arm studies for the indication. These results suggest that historical success rates may reflect not only the important features of trials, which can be observed directly from study design and results, but also the background status of trials in clinical development pathways.
Collapse
Affiliation(s)
- Can Wu
- Laboratory of Pharmaceutical Regulatory Science, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shunsuke Ono
- Laboratory of Pharmaceutical Regulatory Science, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
98
|
Tang J, Wang Y, Luo Y, Fu J, Zhang Y, Li Y, Xiao Z, Lou Y, Qiu Y, Zhu F. Computational advances of tumor marker selection and sample classification in cancer proteomics. Comput Struct Biotechnol J 2020; 18:2012-2025. [PMID: 32802273 PMCID: PMC7403885 DOI: 10.1016/j.csbj.2020.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer proteomics has become a powerful technique for characterizing the protein markers driving transformation of malignancy, tracing proteome variation triggered by therapeutics, and discovering the novel targets and drugs for the treatment of oncologic diseases. To facilitate cancer diagnosis/prognosis and accelerate drug target discovery, a variety of methods for tumor marker identification and sample classification have been developed and successfully applied to cancer proteomic studies. This review article describes the most recent advances in those various approaches together with their current applications in cancer-related studies. Firstly, a number of popular feature selection methods are overviewed with objective evaluation on their advantages and disadvantages. Secondly, these methods are grouped into three major classes based on their underlying algorithms. Finally, a variety of sample separation algorithms are discussed. This review provides a comprehensive overview of the advances on tumor maker identification and patients/samples/tissues separations, which could be guidance to the researches in cancer proteomics.
Collapse
Key Words
- ANN, Artificial Neural Network
- ANOVA, Analysis of Variance
- CFS, Correlation-based Feature Selection
- Cancer proteomics
- Computational methods
- DAPC, Discriminant Analysis of Principal Component
- DT, Decision Trees
- EDA, Estimation of Distribution Algorithm
- FC, Fold Change
- GA, Genetic Algorithms
- GR, Gain Ratio
- HC, Hill Climbing
- HCA, Hierarchical Cluster Analysis
- IG, Information Gain
- LDA, Linear Discriminant Analysis
- LIMMA, Linear Models for Microarray Data
- MBF, Markov Blanket Filter
- MWW, Mann–Whitney–Wilcoxon test
- OPLS-DA, Orthogonal Partial Least Squares Discriminant Analysis
- PCA, Principal Component Analysis
- PLS-DA, Partial Least Square Discriminant Analysis
- RF, Random Forest
- RF-RFE, Random Forest with Recursive Feature Elimination
- SA, Simulated Annealing
- SAM, Significance Analysis of Microarrays
- SBE, Sequential Backward Elimination
- SFS, and Sequential Forward Selection
- SOM, Self-organizing Map
- SU, Symmetrical Uncertainty
- SVM, Support Vector Machine
- SVM-RFE, Support Vector Machine with Recursive Feature Elimination
- Sample classification
- Tumor marker selection
- sPLSDA, Sparse Partial Least Squares Discriminant Analysis
- t-SNE, Student t Distribution
- χ2, Chi-square
Collapse
Affiliation(s)
- Jing Tang
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Yi Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziyu Xiao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Yunqing Qiu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Feng Zhu
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
99
|
Yang Q, Wang Y, Zhang Y, Li F, Xia W, Zhou Y, Qiu Y, Li H, Zhu F. NOREVA: enhanced normalization and evaluation of time-course and multi-class metabolomic data. Nucleic Acids Res 2020; 48:W436-W448. [PMID: 32324219 PMCID: PMC7319444 DOI: 10.1093/nar/gkaa258] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/21/2020] [Accepted: 04/04/2020] [Indexed: 12/23/2022] Open
Abstract
Biological processes (like microbial growth & physiological response) are usually dynamic and require the monitoring of metabolic variation at different time-points. Moreover, there is clear shift from case-control (N=2) study to multi-class (N>2) problem in current metabolomics, which is crucial for revealing the mechanisms underlying certain physiological process, disease metastasis, etc. These time-course and multi-class metabolomics have attracted great attention, and data normalization is essential for removing unwanted biological/experimental variations in these studies. However, no tool (including NOREVA 1.0 focusing only on case-control studies) is available for effectively assessing the performance of normalization method on time-course/multi-class metabolomic data. Thus, NOREVA was updated to version 2.0 by (i) realizing normalization and evaluation of both time-course and multi-class metabolomic data, (ii) integrating 144 normalization methods of a recently proposed combination strategy and (iii) identifying the well-performing methods by comprehensively assessing the largest set of normalizations (168 in total, significantly larger than those 24 in NOREVA 1.0). The significance of this update was extensively validated by case studies on benchmark datasets. All in all, NOREVA 2.0 is distinguished for its capability in identifying well-performing normalization method(s) for time-course and multi-class metabolomics, which makes it an indispensable complement to other available tools. NOREVA can be accessed at https://idrblab.org/noreva/.
Collapse
Affiliation(s)
- Qingxia Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weiqi Xia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation & The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Yunqing Qiu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation & The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Honglin Li
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
100
|
Wang C, Zhao N, Sun K, Zhang Y. A Cancer Gene Module Mining Method Based on Bio-Network of Multi-Omics Gene Groups. Front Oncol 2020; 10:1159. [PMID: 32637361 PMCID: PMC7317001 DOI: 10.3389/fonc.2020.01159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/08/2020] [Indexed: 11/13/2022] Open
Abstract
The initiation, promotion and progression of cancer are highly associated to the environment a human lives in as well as individual genetic factors. In view of the dangers to life and health caused by this abnormally complex systemic disease, many top scientific research institutions around the world have been actively carrying out research in order to discover the pathogenic mechanisms driving cancer occurrence and development. The emergence of high-throughput sequencing technology has greatly advanced oncology research and given rise to the revelation of important oncogenes and the interrelationship among them. Here, we have studied heterogeneous multi-level data within a context of integrated data, and scientifically introduced lncRNA omics data to construct multi-omics bio-network models, allowing the screening of key cancer-related gene groups. We propose a compactness clustering algorithm based on corrected cumulative rank scores, which uses the functional similarity between groups of genes as a distance measure to excavate key gene modules for abnormal regulation contained in gene groups through clustering. We also conducted a survival analysis using our results and found that our model could divide groups of different levels very well. The results also demonstrate that the integration of multi-omics biological data, key gene modules and their dysregulated gene groups can be discovered, which is crucial for cancer research.
Collapse
Affiliation(s)
- Chunyu Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ning Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kai Sun
- Thoracic Surgery Department, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin, China
| | - Ying Zhang
- Department of Pharmacy, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin, China
| |
Collapse
|