1
|
Zhang W, Wu Y, Yuan Y, Wang L, Yu B, Li X, Yao Z, Liang B. Identification of key biomarkers for predicting atherosclerosis progression in polycystic ovary syndrome via bioinformatics analysis and machine learning. Comput Biol Med 2024; 183:109239. [PMID: 39396400 DOI: 10.1016/j.compbiomed.2024.109239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is one of the most significant cardiovascular risk factors, playing vital roles in various cardiovascular diseases such as atherosclerosis (AS). This study attempted to explore key biomarkers for predicting AS in patients with PCOS and to investigate the role of immune cell infiltration in this process. METHODS We downloaded the expression matrix of AS (GSE100927, GSE28829) and PCOS (GSE54248) from the Gene Expression Omnibus (GEO) database. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were used to identify PCOS-related genes in AS. Functional enrichment analysis was employed to reveal underlying mechanisms. Then, Protein-protein interaction (PPI) and three machine learning algorithms were used to screen the hub genes, including the Least Absolute Shrinkage and Selection Operator (LASSO), Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and Random Forest (RF). Moreover, the receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA) were applied to evaluate the diagnostic value of the nomogram model. Finally, we performed immune cell infiltration and single-gene GSEA. RESULTS A total of 41 genes were identified as PCOS-related genes in AS, with functional analysis indicating that the potential pathogenesis lies in inflammatory and immune responses. Furthermore, we identified two hub genes (MMP9 and P2RY13) by three machine learning algorithms. The nomogram model based on MMP9 and P2RY13 can be used as a new diagnostic model to differentiate AS in PCOS women (AUC>0.9). The calibration curves and DCA curves demonstrated the excellent discriminative ability and clinical practicality of this nomogram. Finally, immune infiltration analysis revealed the disorder of immunocytes in AS. The two gene expressions were negatively correlated with Monocyte and Macrophages M1, while positively correlated with Macrophages M0. Single gene GSEA analysis suggested that the MMP9 and P2RY13 might be involved in the metabolism and inflammation responses. CONCLUSION We identified MMP9 and P2RY13 as the biomarkers and developed a new nomogram for early diagnosing AS based on them in PCOS patients. Our findings may provide new insights into the diagnosis, prevention, and treatment targets of PCOS-associated AS.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Yalin Wu
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Yalin Yuan
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Leigang Wang
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Bing Yu
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Xin Li
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Zhong Yao
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Bin Liang
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China.
| |
Collapse
|
2
|
Shen X, Zhang Y, Li J, Zhou Y, Butensky SD, Zhang Y, Cai Z, DeWan AT, Khan SA, Yan H, Johnson CH, Zhu F. OncoSexome: the landscape of sex-based differences in oncologic diseases. Nucleic Acids Res 2024:gkae1003. [PMID: 39535034 DOI: 10.1093/nar/gkae1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/28/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The NIH policy on sex as biological variable (SABV) emphasized the importance of sex-based differences in precision oncology. Over 50% of clinically actionable oncology genes are sex-biased, indicating differences in drug efficacy. Research has identified sex differences in non-reproductive cancers, highlighting the need for comprehensive sex-based cancer data. We therefore developed OncoSexome, a multidimensional knowledge base describing sex-based differences in cancer (https://idrblab.org/OncoSexome/) across four key topics: antineoplastic drugs and responses (SDR), oncology-related biomarkers (SBM), risk factors (SRF) and microbial landscape (SML). SDR covers sex-based differences in 2051 anticancer drugs; SBM describes 12 551 sex-differential biomarkers; SRF illustrates 350 sex-dependent risk factors; SML demonstrates 1386 microbes with sex-differential abundances associated with cancer development. OncoSexome is unique in illuminating multifaceted influences of biological sex on cancer, providing both external and endogenous contributors to cancer development and describing sex-based differences for the broadest oncological classes. Given the increasing global research interest in sex-based differences, OncoSexome is expected to impact future precision oncology practices significantly.
Collapse
Affiliation(s)
- Xinyi Shen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Jiamin Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | | | - Yechi Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
- School of Public Health, Zhejiang University, Hangzhou 310058, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Sajid A Khan
- Yale School of Medicine, Yale University, New Haven 06510, USA
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven 06510, USA
| | - Hong Yan
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, 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
|
3
|
Harati Kabir V, Mahdavifar Khayati R, Motie Nasrabadi A, Nabavi SM. Prediction of Expanded Disability Status Scale in patients with MS using deep learning. Comput Biol Med 2024; 182:109143. [PMID: 39270459 DOI: 10.1016/j.compbiomed.2024.109143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
Multiple sclerosis (MS) is a chronic neurological condition that leads to significant disability in patients. Accurate prediction of disease progression, specifically the Expanded Disability Status Scale (EDSS), is crucial for personalizing treatment and improving patient outcomes. This study aims to develop a robust deep neural network framework to predict EDSS in MS patients using MRI scans. Our model demonstrates high accuracy and reliability in both lesion segmentation and disability classification tasks. For segmentation, the model achieves a Dice Coefficient of 0.87, a Jaccard Index of 0.79, sensitivity of 0.85, and specificity of 0.88. In classification, it attains an overall accuracy of 91.2 %, with a precision of 0.89, recall of 0.88, and an F1-Score of 0.885. Ablation studies highlight the significant impact of integrating T2-weighted and FLAIR images, improving accuracy from 85.7 % (T1-weighted alone) to 93.4 %. Comparative analysis with state-of-the-art methods demonstrates our model's superiority, outperforming Method A and Method B in both accuracy and F1-Score. Despite these advancements, challenges such as data quality, sample size, and computational complexity remain. Future research should focus on standardizing imaging protocols, incorporating larger and more diverse datasets, and optimizing model efficiency. Advancing deep learning architectures and utilizing multimodal data can enhance predictive power and facilitate real-time clinical applications. Our study significantly contributes to refining MS treatment strategies by providing a comprehensive evaluation of our model's performance and addressing key limitations. Accurate disability predictions enable personalized treatments, early interventions, and improved patient outcomes, thus enhancing the quality of life for individuals affected by MS.
Collapse
Affiliation(s)
| | | | | | - Seyed Massood Nabavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| |
Collapse
|
4
|
Li Y, Li F, Duan Z, Liu R, Jiao W, Wu H, Zhu F, Xue W. SYNBIP 2.0: epitopes mapping, sequence expansion and scaffolds discovery for synthetic binding protein innovation. Nucleic Acids Res 2024:gkae893. [PMID: 39413165 DOI: 10.1093/nar/gkae893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/18/2024] Open
Abstract
Synthetic binding proteins (SBPs) represent a pivotal class of artificially engineered proteins, meticulously crafted to exhibit targeted binding properties and specific functions. Here, the SYNBIP database, a comprehensive resource for SBPs, has been significantly updated. These enhancements include (i) featuring 3D structures of 899 SBP-target complexes to illustrate the binding epitopes of SBPs, (ii) using the structures of SBPs in the monomer or complex forms with target proteins, their sequence space has been expanded five times to 12 025 by integrating a structure-based protein generation framework and a protein property prediction tool, (iii) offering detailed information on 78 473 newly identified SBP-like scaffolds from the RCSB Protein Data Bank, and an additional 16 401 555 ones from the AlphaFold Protein Structure Database, and (iv) the database is regularly updated, incorporating 153 new SBPs. Furthermore, the structural models of all SBPs have been enhanced through the application of the AlphaFold2, with their clinical statuses concurrently refreshed. Additionally, the design methods employed for each SBP are now prominently featured in the database. In sum, SYNBIP 2.0 is designed to provide researchers with essential SBP data, facilitating their innovation in research, diagnosis and therapy. SYNBIP 2.0 is now freely accessible at https://idrblab.org/synbip/.
Collapse
Affiliation(s)
- Yanlin Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Fengcheng Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, Zhejiang 310052, China
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Zixin Duan
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Ruihan Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Wantong Jiao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Haibo Wu
- School of Life Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Weiwei Xue
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| |
Collapse
|
5
|
Zhang Y, Lian X, Xu H, Zhu S, Zhang H, Ni Z, Fu T, Liu S, Tao L, Zhou Y, Zhu F. OrgXenomics: an integrated proteomic knowledge base for patient-derived organoid and xenograft. Nucleic Acids Res 2024:gkae861. [PMID: 39373514 DOI: 10.1093/nar/gkae861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Patient-derived models (PDMs, particularly organoids and xenografts) are irreplaceable tools for precision medicine, from target development to lead identification, then to preclinical evaluation, and finally to clinical decision-making. So far, PDM-based proteomics has emerged to be one of the cutting-edge directions and massive data have been accumulated. However, such PDM-based proteomic data have not been provided by any of the available databases, and proteomics profiles of all proteins in proteomic study are also completely absent from existing databases. Herein, an integrated database named 'OrgXenomics' was thus developed to provide the proteomic data for PDMs, which was unique in (a) explicitly describing the establishment detail for a wide array of models, (b) systematically providing the proteomic profiles (expression/function/interaction) for all proteins in studied proteomic analysis and (c) comprehensively giving the raw data for diverse organoid/xenograft-based proteomic studies of various diseases. Our OrgXenomics was expected to server as one good complement to existing proteomic databases, and had great implication for the practice of precision medicine, which could be accessed at: https://idrblab.org/orgxenomics/.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, 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
| | - Xichen Lian
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hangwei Xu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Sisi Zhu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Zhang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, 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
|
6
|
Zhang Y, Jiang W, Li T, Xu H, Zhu Y, Fang K, Ren X, Wang S, Chen Y, Zhou Y, Zhu F. SubCELL: the landscape of subcellular compartment-specific molecular interactions. Nucleic Acids Res 2024:gkae863. [PMID: 39373488 DOI: 10.1093/nar/gkae863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
The subcellular compartment-specific molecular interactions (SCSIs) are the building blocks for most molecular functions, biological processes and disease pathogeneses. Extensive experiments have therefore been conducted to accumulate the valuable information of SCSIs, but none of the available databases has been constructed to describe those data. In this study, a novel knowledge base SubCELL is thus introduced to depict the landscape of SCSIs among DNAs/RNAs/proteins. This database is UNIQUE in (a) providing, for the first time, the experimentally-identified SCSIs, (b) systematically illustrating a large number of SCSIs inferred based on well-established method and (c) collecting experimentally-determined subcellular locations for the DNAs/RNAs/proteins of diverse species. Given the essential physiological/pathological role of SCSIs, the SubCELL is highly expected to have great implications for modern molecular biological study, which can be freely accessed with no login requirement at: https://idrblab.org/subcell/.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, 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
| | - Wanghao Jiang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Teng Li
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hangwei Xu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yimiao Zhu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Kerui Fang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Ren
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shanshan Wang
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Yuzong Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, 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
|
7
|
Sun X, Li H, Chen Z, Zhang Y, Wei Z, Xu H, Liao Y, Jiang W, Ge Y, Zheng L, Li T, Wu Y, Luo M, Fang L, Dong X, Xiao M, Han L, Jia Q, Zhu F. PDCdb: the biological activity and pharmaceutical information of peptide-drug conjugate (PDC). Nucleic Acids Res 2024:gkae859. [PMID: 39360619 DOI: 10.1093/nar/gkae859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Peptide-drug conjugates (PDCs) have emerged as a promising class of targeted therapeutics with substantial pharmaceutical advantages and market potentials, which is a combination of a peptide (selective to the disease-relevant target), a linker (stable in circulation but cleavable at target site) and a cytotoxic/radioactive drug (efficacious/traceable for disease). Among existing PDCs, those based on radiopharmaceuticals (a.k.a. radioactive drugs) are valued due to their accurate imaging and targeted destruction of disease sites. It's demanded to accumulate the biological activity and pharmaceutical information of PDCs. Herein, a database PDCdb was thus constructed to systematically describe these valuable data. Particularly, biological activities for 2036 PDCs were retrieved from literatures, which resulted in 1684, 613 and 2753 activity data generated based on clinical trial, animal model and cell line, respectively. Furthermore, the pharmaceutical information for all 2036 PDCs was collected, which gave the diverse data of (a) ADME property, plasma half-life and administration approach of a PDC and (b) chemical modification, primary target, mode of action, conjugating feature of the constituent peptide/linker/drug. In sum, PDCdb systematically provided the biological activities and pharmaceutical information for the most comprehensive list of PDCs among the available databases, which was expected to attract broad interest from related communities and could be freely accessible at: https://idrblab.org/PDCdb/.
Collapse
Affiliation(s)
- Xiuna Sun
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hanyang Li
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhangle Wei
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hangwei Xu
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yang Liao
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Wanghao Jiang
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yichao Ge
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Lingyan Zheng
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Teng Li
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yuting Wu
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Meiyin Luo
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Luo Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310005, China
| | - Xiaowu Dong
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Mang Xiao
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Lianyi Han
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai 315211, China
| | - Qingzhong Jia
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Feng Zhu
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
- College of Pharmaceutical Sciences, Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
8
|
Esfandiari A, Nasiri N. Gene selection and cancer classification using interaction-based feature clustering and improved-binary Bat algorithm. Comput Biol Med 2024; 181:109071. [PMID: 39205342 DOI: 10.1016/j.compbiomed.2024.109071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
In high-dimensional gene expression data, selecting an optimal subset of genes is crucial for achieving high classification accuracy and reliable diagnosis of diseases. This paper proposes a two-stage hybrid model for gene selection based on clustering and a swarm intelligence algorithm to identify the most informative genes with high accuracy. First, a clustering-based multivariate filter approach is performed to explore the interactions between the features and eliminate any redundant or irrelevant ones. Then, by controlling for the problem of premature convergence in the binary Bat algorithm, the optimal gene subset is determined using different classifiers with the Monte Carlo cross-validation data partitioning model. The effectiveness of our proposed framework is evaluated using eight gene expression datasets, by comparison with other recently published algorithms in the literature. Experiments confirm that in seven out of eight datasets, the proposed method can achieve superior results in terms of classification accuracy and gene subset size. In particular, it achieves a classification accuracy of 100% in Lymphoma and Ovarian datasets and above 97.4% in the rest with a minimum number of genes. The results demonstrate that our proposed algorithm has the potential to solve the feature selection problem in different applications with high-dimensional datasets.
Collapse
Affiliation(s)
- Ahmad Esfandiari
- Department of Computer Engineering, Sari Branch, Islamic Azad University, Sari, Iran.
| | - Niki Nasiri
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
9
|
Li F, Mou M, Li X, Xu W, Yin J, Zhang Y, Zhu F. DrugMAP 2.0: molecular atlas and pharma-information of all drugs. Nucleic Acids Res 2024:gkae791. [PMID: 39271119 DOI: 10.1093/nar/gkae791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
The escalating costs and high failure rates have decelerated the pace of drug development, which amplifies the research interests in developing combinatorial/repurposed drugs and understanding off-target adverse drug reaction (ADR). In other words, it is demanded to delineate the molecular atlas and pharma-information for the combinatorial/repurposed drugs and off-target interactions. However, such invaluable data were inadequately covered by existing databases. In this study, a major update was thus conducted to the DrugMAP, which accumulated (a) 20831 combinatorial drugs and their interacting atlas involving 1583 pharmacologically important molecules; (b) 842 repurposed drugs and their interacting atlas with 795 molecules; (c) 3260 off-targets relevant to the ADRs of 2731 drugs and (d) various types of pharmaceutical information, including diverse ADMET properties, versatile diseases, and various ADRs/off-targets. With the growing demands for discovering combinatorial/repurposed therapies and the rapidly emerging interest in AI-based drug discovery, DrugMAP was highly expected to act as an indispensable supplement to existing databases facilitating drug discovery, which was accessible at: https://idrblab.org/drugmap/.
Collapse
Affiliation(s)
- Fengcheng Li
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
- State Key Lab of Advanced Drug Delivery and Release Systems, 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
| | - Xiaoyi Li
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
| | - Weize Xu
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
| | - Jiayi Yin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
- State Key Lab of Advanced Drug Delivery and Release Systems, 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
|
10
|
Teo YX, Lee RE, Nurzaman SG, Tan CP, Chan PY. Action tremor features discovery for essential tremor and Parkinson's disease with explainable multilayer BiLSTM. Comput Biol Med 2024; 180:108957. [PMID: 39098236 DOI: 10.1016/j.compbiomed.2024.108957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/04/2024] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
The tremors of Parkinson's disease (PD) and essential tremor (ET) are known to have overlapping characteristics that make it complicated for clinicians to distinguish them. While deep learning is robust in detecting features unnoticeable to humans, an opaque trained model is impractical in clinical scenarios as coincidental correlations in the training data may be used by the model to make classifications, which may result in misdiagnosis. This work aims to overcome the aforementioned challenge of deep learning models by introducing a multilayer BiLSTM network with explainable AI (XAI) that can better explain tremulous characteristics and quantify the respective discovered important regions in tremor differentiation. The proposed network classifies PD, ET, and normal tremors during drinking actions and derives the contribution from tremor characteristics, (i.e., time, frequency, amplitude, and actions) utilized in the classification task. The analysis shows that the XAI-BiLSTM marks the regions with high tremor amplitude as important in classification, which is verified by a high correlation between relevance distribution and tremor displacement amplitude. The XAI-BiLSTM discovered that the transition phases from arm resting to lifting (during the drinking cycle) is the most important action to classify tremors. Additionally, the XAI-BiLSTM reveals frequency ranges that only contribute to the classification of one tremor class, which may be the potential distinctive feature to overcome the overlapping frequencies problem. By revealing critical timing and frequency patterns unique to PD and ET tremors, this proposed XAI-BiLSTM model enables clinicians to make more informed classifications, potentially reducing misclassification rates and improving treatment outcomes.
Collapse
Affiliation(s)
- Yu Xuan Teo
- Department of Electrical & Robotics Engineering, School of Engineering, Monash University Malaysia, Malaysia.
| | - Rui En Lee
- Department of Electrical & Robotics Engineering, School of Engineering, Monash University Malaysia, Malaysia.
| | - Surya Girinatha Nurzaman
- Department of Mechanical Engineering, School of Engineering, Monash University Malaysia, Bandar Sunway, Malaysia.
| | - Chee Pin Tan
- Department of Electrical & Robotics Engineering, School of Engineering, Monash University Malaysia, Malaysia.
| | - Ping Yi Chan
- Department of Electrical & Robotics Engineering, School of Engineering, Monash University Malaysia, Malaysia.
| |
Collapse
|
11
|
Alkadri S, Del Maestro RF, Driscoll M. Unveiling surgical expertise through machine learning in a novel VR/AR spinal simulator: A multilayered approach using transfer learning and connection weights analysis. Comput Biol Med 2024; 179:108809. [PMID: 38944904 DOI: 10.1016/j.compbiomed.2024.108809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Virtual and augmented reality surgical simulators, integrated with machine learning, are becoming essential for training psychomotor skills, and analyzing surgical performance. Despite the promise of methods like the Connection Weights Algorithm, the small sample sizes (small number of participants (N)) typical of these trials challenge the generalizability and robustness of models. Approaches like data augmentation and transfer learning from models trained on similar surgical tasks address these limitations. OBJECTIVE To demonstrate the efficacy of artificial neural network and transfer learning algorithms in evaluating virtual surgical performances, applied to a simulated oblique lateral lumbar interbody fusion technique in an augmented and virtual reality simulator. DESIGN The study developed and integrated artificial neural network algorithms within a novel simulator platform, using data from the simulated tasks to generate 276 performance metrics across motion, safety, and efficiency. Innovatively, it applies transfer learning from a pre-trained ANN model developed for a similar spinal simulator, enhancing the training process, and addressing the challenge of small datasets. SETTING Musculoskeletal Biomechanics Research Lab; Neurosurgical Simulation and Artificial Intelligence Learning Centre, McGill University, Montreal, Canada. PARTICIPANTS Twenty-seven participants divided into 3 groups: 9 post-residents, 6 senior and 12 junior residents. RESULTS Two models, a stand-alone model trained from scratch and another leveraging transfer learning, were trained on nine selected surgical metrics achieving 75 % and 87.5 % testing accuracy respectively. CONCLUSIONS This study presents a novel blueprint for addressing limited datasets in surgical simulations through the strategic use of transfer learning and data augmentation. It also evaluates and reinforces the application of the Connection Weights Algorithm from our previous publication. Together, these methodologies not only enhance the precision of performance classification but also advance the validation of surgical training platforms.
Collapse
Affiliation(s)
- Sami Alkadri
- Musculoskeletal Biomechanics Research Lab, Department of Mechanical Engineering, McGill University, Macdonald Engineering Building, 815 Sherbrooke St W, Montreal, H3A 2K7, QC, Canada; Neurosurgical Simulation and Artificial Intelligence Learning Centre, Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, 2200 Leo Pariseau, Suite, 2210, Montreal, H2X 4B3, Quebec, Canada
| | - Rolando F Del Maestro
- Neurosurgical Simulation and Artificial Intelligence Learning Centre, Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, 2200 Leo Pariseau, Suite, 2210, Montreal, H2X 4B3, Quebec, Canada
| | - Mark Driscoll
- Musculoskeletal Biomechanics Research Lab, Department of Mechanical Engineering, McGill University, Macdonald Engineering Building, 815 Sherbrooke St W, Montreal, H3A 2K7, QC, Canada; Orthopaedic Research Lab, Montreal General Hospital, 1650 Cedar Ave (LS1.409), Montreal, H3G 1A4, Quebec, Canada.
| |
Collapse
|
12
|
Qiu W, Zhang S, Yu W, Liu J, Wu H. Non-coding RNAs in hepatocellular carcinoma metastasis: Remarkable indicators and potential oncogenic mechanism. Comput Biol Med 2024; 180:108867. [PMID: 39089114 DOI: 10.1016/j.compbiomed.2024.108867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 06/12/2024] [Accepted: 07/07/2024] [Indexed: 08/03/2024]
Abstract
Non-coding RNAs (ncRNAs), as key regulators involving in intercellular biological processes, are more prominent in many malignancies, especially for hepatocellular carcinoma (HCC). Herein, we conduct a comprehensive review to summarize diverse ncRNAs roles in HCC metastatic mechanism. We focus on four signaling pathways that predominate in HCC metastatic process, including Wnt/β-catenin, HIF-1α, IL-6, and TGF-β pathways. MicroRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) employed different mechanisms to participate in the regulation of the key genes in these pathways, typical as interaction with DNA to control transcription, with RNA to control translation, and with protein to control stability. Therefore, ncRNAs may become potential biomarkers and therapeutic targets for HCC metastasis.
Collapse
Affiliation(s)
- Wenqi Qiu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Song Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huiling Wu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Singh S, Kaur N, Gehlot A. Application of artificial intelligence in drug design: A review. Comput Biol Med 2024; 179:108810. [PMID: 38991316 DOI: 10.1016/j.compbiomed.2024.108810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/31/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Artificial intelligence (AI) is a field of computer science that involves acquiring information, developing rule bases, and mimicking human behaviour. The fundamental concept behind AI is to create intelligent computer systems that can operate with minimal human intervention or without any intervention at all. These rule-based systems are developed using various machine learning and deep learning models, enabling them to solve complex problems. AI is integrated with these models to learn, understand, and analyse provided data. The rapid advancement of Artificial Intelligence (AI) is reshaping numerous industries, with the pharmaceutical sector experiencing a notable transformation. AI is increasingly being employed to automate, optimize, and personalize various facets of the pharmaceutical industry, particularly in pharmacological research. Traditional drug development methods areknown for being time-consuming, expensive, and less efficient, often taking around a decade and costing billions of dollars. The integration of artificial intelligence (AI) techniques addresses these challenges by enabling the examination of compounds with desired properties from a vast pool of input drugs. Furthermore, it plays a crucial role in drug screening by predicting toxicity, bioactivity, ADME properties (absorption, distribution, metabolism, and excretion), physicochemical properties, and more. AI enhances the drug design process by improving the efficiency and accuracy of predicting drug behaviour, interactions, and properties. These approaches further significantly improve the precision of drug discovery processes and decrease clinical trial costs leading to the development of more effective drugs.
Collapse
Affiliation(s)
- Simrandeep Singh
- Department of Electronics & Communication Engineering, UCRD, Chandigarh University, Gharuan, Punjab, India.
| | - Navjot Kaur
- Department of Pharmacognosy, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, India
| | - Anita Gehlot
- Uttaranchal Institute of technology, Uttaranchal University, Dehradun, India
| |
Collapse
|
14
|
Maryam, Rehman MU, Hussain I, Tayara H, Chong KT. A graph neural network approach for predicting drug susceptibility in the human microbiome. Comput Biol Med 2024; 179:108729. [PMID: 38955124 DOI: 10.1016/j.compbiomed.2024.108729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 07/04/2024]
Abstract
Recent studies have illuminated the critical role of the human microbiome in maintaining health and influencing the pharmacological responses of drugs. Clinical trials, encompassing approximately 150 drugs, have unveiled interactions with the gastrointestinal microbiome, resulting in the conversion of these drugs into inactive metabolites. It is imperative to explore the field of pharmacomicrobiomics during the early stages of drug discovery, prior to clinical trials. To achieve this, the utilization of machine learning and deep learning models is highly desirable. In this study, we have proposed graph-based neural network models, namely GCN, GAT, and GINCOV models, utilizing the SMILES dataset of drug microbiome. Our primary objective was to classify the susceptibility of drugs to depletion by gut microbiota. Our results indicate that the GINCOV surpassed the other models, achieving impressive performance metrics, with an accuracy of 93% on the test dataset. This proposed Graph Neural Network (GNN) model offers a rapid and efficient method for screening drugs susceptible to gut microbiota depletion and also encourages the improvement of patient-specific dosage responses and formulations.
Collapse
Affiliation(s)
- Maryam
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Mobeen Ur Rehman
- Khalifa University Center for Autonomous Robotic Systems (KUCARS), Khalifa University, United Arab Emirates
| | - Irfan Hussain
- Khalifa University Center for Autonomous Robotic Systems (KUCARS), Khalifa University, United Arab Emirates
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju, 54896, South Korea.
| | - Kil To Chong
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, South Korea; Advances Electronics and Information Research Centre, Jeonbuk National University, Jeonju, 54896, South Korea.
| |
Collapse
|
15
|
Nguyen-Vo TH, Do TTT, Nguyen BP. Multitask Learning on Graph Convolutional Residual Neural Networks for Screening of Multitarget Anticancer Compounds. J Chem Inf Model 2024. [PMID: 39197175 DOI: 10.1021/acs.jcim.4c00643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Recently, various modern experimental screening pipelines and assays have been developed to find promising anticancer drug candidates. However, it is time-consuming and almost infeasible to screen an immense number of compounds for anticancer activity via experimental approaches. To partially address this issue, several computational advances have been proposed. In this study, we present iACP-GCR, a model based on multitask learning on graph convolutional residual neural networks with two types of shortcut connections, to identify multitarget anticancer compounds. In our architecture, the graph convolutional residual neural networks are shared by all the prediction tasks before being separately customized. The NCI-60 data set, one of the most reliable and well-known sources of experimentally verified compounds, was used to develop our model. From that data set, we collected and refined data about compounds screened across nine cancer types (panels), including breast, central nervous system, colon, leukemia, nonsmall cell lung, melanoma, ovarian, prostate, and renal, for model training and evaluation. The model performance evaluated on an independent test set shows that iACP-GCR surpasses the three advanced computational methods for multitask learning. The integration of two shortcut connection types in the shared networks also improves the prediction efficiency. We also deployed the model as a public web server to assist the research community in screening potential anticancer compounds.
Collapse
Affiliation(s)
- Thanh-Hoang Nguyen-Vo
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Trang T T Do
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Binh P Nguyen
- Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand
| |
Collapse
|
16
|
Zheng X, Lamoth CJ, Timmerman H, Otten E, Reneman MF. Establishing central sensitization inventory cut-off values in Dutch-speaking patients with chronic low back pain by unsupervised machine learning. Comput Biol Med 2024; 178:108739. [PMID: 38875910 DOI: 10.1016/j.compbiomed.2024.108739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/29/2024] [Accepted: 06/08/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Human Assumed Central Sensitization (HACS) is involved in the development and maintenance of chronic low back pain (CLBP). The Central Sensitization Inventory (CSI) was developed to evaluate the presence of HACS, with a cut-off value of 40/100. However, various factors including pain conditions (e.g., CLBP), contexts, and gender may influence this cut-off value. Unsupervised clustering approaches can address these complexities by considering diverse factors and exploring possible HACS-related subgroups. Therefore, this study aimed to determine the cut-off values for a Dutch-speaking population with CLBP based on unsupervised machine learning. METHODS Questionnaire data covering pain, physical, and psychological aspects were collected from patients with CLBP and aged-matched healthy controls (HC). Four clustering approaches were applied to identify HACS-related subgroups based on the questionnaire data and gender. The clustering performance was assessed using internal and external indicators. Subsequently, receiver operating characteristic (ROC) analysis was conducted on the best clustering results to determine the optimal cut-off values. RESULTS The study included 63 HCs and 88 patients with CLBP. Hierarchical clustering yielded the best results, identifying three clusters: healthy group, CLBP with low HACS level, and CLBP with high HACS level groups. The cut-off value for the overall groups were 35 (sensitivity 0.76, specificity 0.76). CONCLUSION This study found distinct patient subgroups. An overall CSI cut-off value of 35 was suggested. This study may provide new insights into identifying HACS-related patterns and contributes to establishing accurate cut-off values.
Collapse
Affiliation(s)
- Xiaoping Zheng
- University of Groningen, University Medical Center Groningen, Department of Human Movement Sciences, Groningen, the Netherlands
| | - Claudine Jc Lamoth
- University of Groningen, University Medical Center Groningen, Department of Human Movement Sciences, Groningen, the Netherlands
| | - Hans Timmerman
- University of Groningen, University Medical Center Groningen, Department of Anesthesiology, Pain Center, Groningen, the Netherlands
| | - Egbert Otten
- University of Groningen, University Medical Center Groningen, Department of Human Movement Sciences, Groningen, the Netherlands
| | - Michiel F Reneman
- University of Groningen, University Medical Center Groningen, Department of Rehabilitation Medicine, Groningen, the Netherlands.
| |
Collapse
|
17
|
Srisongkram T. DeepRA: A novel deep learning-read-across framework and its application in non-sugar sweeteners mutagenicity prediction. Comput Biol Med 2024; 178:108731. [PMID: 38870727 DOI: 10.1016/j.compbiomed.2024.108731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/07/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024]
Abstract
Non-sugar sweeteners (NSSs) or artificial sweeteners have long been used as food chemicals since World War II. NSSs, however, also raise a concern about their mutagenicity. Evaluating the mutagenic ability of NSSs is crucial for food safety; this step is needed for every new chemical registration in the food and pharmaceutical industries. A computational assessment provides less time, money, and involved animals than the in vivo experiments; thus, this study developed a novel computational method from an ensemble convolutional deep neural network and read-across algorithms, called DeepRA, to classify the mutagenicity of chemicals. The mutagenicity data were obtained from the curated Ames test data set. The DeepRA model was developed using both molecular descriptors and molecular fingerprints. The obtained DeepRA model provides accurate and reliable mutagenicity classification through an independent test set. This model was then used to examine the NSSs-related chemicals, enabling the evaluation of mutagenicity from the NSSs-like substances. Finally, this model was publicly available at https://github.com/taraponglab/deepra for further use in chemical regulation and risk assessment.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand.
| |
Collapse
|
18
|
Chowdhury FA, Hosain MK, Bin Islam MS, Hossain MS, Basak P, Mahmud S, Murugappan M, Chowdhury MEH. ECG waveform generation from radar signals: A deep learning perspective. Comput Biol Med 2024; 176:108555. [PMID: 38749323 DOI: 10.1016/j.compbiomed.2024.108555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/01/2024] [Accepted: 05/05/2024] [Indexed: 05/31/2024]
Abstract
Cardiovascular diagnostics relies heavily on the ECG (ECG), which reveals significant information about heart rhythm and function. Despite their significance, traditional ECG measures employing electrodes have limitations. As a result of extended electrode attachments, patients may experience skin irritation or pain, and motion artifacts may interfere with signal accuracy. Additionally, ECG monitoring usually requires highly trained professionals and specialized equipment, which increases the treatment's complexity and cost. In critical care scenarios, such as continuous monitoring of hospitalized patients, wearable sensors for collecting ECG data may be difficult to use. Although there are issues with ECG, it remains a valuable tool for diagnosing and monitoring cardiac disorders due to its non-invasive nature and the detailed information it provides about the heart. The goal of this study is to present an innovative method for generating continuous ECG waveforms from non-contact radar data by using Deep Learning. The method can eliminate the need for invasive or wearable biosensors and expensive equipment to collect ECGs. In this paper, we propose the MultiResLinkNet, a one-dimensional convolutional neural network (1D CNN) model for generating ECG signals from radar waveforms. With the help of a publicly accessible radar benchmark dataset, an end-to-end DL architecture is trained and assessed. There are six ports of raw radar data in this dataset, along with ground truth physiological signals collected from 30 participants in five distinct scenarios: Resting, Valsalva, Apnea, Tilt-up, and Tilt-down. By using strong temporal and spectral measurements, we assessed our proposed framework's ability to convert ECG data from Radar signals in three distinct scenarios, namely Resting, Valsalva, and Apnea (RVA). ECG segmentation performed better by MultiResLinkNet than by state-of-the-art networks in both combined and individual cases. As a result of the simulations, the resting, valsalva, and RVA scenarios showed the highest average temporal values, respectively: 66.09523 ± 19.33, 60.13625 ± 21.92, and 61.86265 ± 21.37. In addition, it exhibited the highest spectral correlation values (82.4388 ± 18.42 (Resting), 77.05186 ± 23.26 (Valsalva), 74.65785 ± 23.17 (Apnea), and 79.96201 ± 20.82 (RVA)), along with minimal temporal and spectral errors in almost every case. The qualitative evaluation revealed strong similarities between generated and actual ECG waveforms. As a result of our method of forecasting ECG patterns from remote radar data, we can monitor high-risk patients, especially those undergoing surgery.
Collapse
Affiliation(s)
- Farhana Ahmed Chowdhury
- Department of Electronics and Telecommunication Engineering, Rajshahi University of Engineering and Technology, Rajshahi, 6204, Bangladesh
| | - Md Kamal Hosain
- Department of Electronics and Telecommunication Engineering, Rajshahi University of Engineering and Technology, Rajshahi, 6204, Bangladesh
| | - Md Sakib Bin Islam
- Department of Biomedical Engineering, Military Institute of Science and Technology, Dhaka, Bangladesh
| | - Md Shafayet Hossain
- Department of Electrical, Electronics and Systems Engineering, Universiti Kebangsaan Malaysia, Bangi, Selangor, 43600, Malaysia
| | - Promit Basak
- Department of Electrical and Electronic Engineering, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Sakib Mahmud
- Department of Electrical Engineering, Qatar University, Doha, 2713, Qatar
| | - M Murugappan
- Intelligent Signal Processing (ISP) Research Lab, Department of Electronics and Communication Engineering, Kuwait College of Science and Technology, Block 4, Doha, Kuwait; Department of Electronics and Communication Engineering, Vels Institute of Sciences, Technology, and Advanced Studies, Chennai, Tamilnadu, India.
| | | |
Collapse
|
19
|
da Silva LSA, Seman LO, Camponogara E, Mariani VC, Dos Santos Coelho L. Bilinear optimization of protein structure prediction: An exact approach via AB off-lattice model. Comput Biol Med 2024; 176:108558. [PMID: 38754216 DOI: 10.1016/j.compbiomed.2024.108558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/25/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
Protein structure prediction (PSP) remains a central challenge in computational biology due to its inherent complexity and high dimensionality. While numerous heuristic approaches have appeared in the literature, their success varies. The AB off-lattice model, which characterizes proteins as sequences of A (hydrophobic) and B (hydrophilic) beads, presents a simplified perspective on PSP. This work presents a mathematical optimization-based methodology capitalizing on the off-lattice AB model. Dissecting the inherent non-linearities of the energy landscape of protein folding allowed for formulating the PSP as a bilinear optimization problem. This formulation was achieved by introducing auxiliary variables and constraints that encapsulate the nuanced relationship between the protein's conformational space and its energy landscape. The proposed bilinear model exhibited notable accuracy in pinpointing the global minimum energy conformations on a benchmark dataset presented by the Protein Data Bank (PDB). Compared to traditional heuristic-based methods, this bilinear approach yielded exact solutions, reducing the likelihood of local minima entrapment. This research highlights the potential of reframing the traditionally non-linear protein structure prediction problem into a bilinear optimization problem through the off-lattice AB model. Such a transformation offers a route toward methodologies that can determine the global solution, challenging current PSP paradigms. Exploration into hybrid models, merging bilinear optimization and heuristic components, might present an avenue for balancing accuracy with computational efficiency.
Collapse
Affiliation(s)
- Luiza Scapinello Aquino da Silva
- Electrical Engineering Graduate Program (PPGEE), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil.
| | - Laio Oriel Seman
- Department of Automation and Systems Engineering, Federal University of Santa Catarina (UFSC), Engenheiro Agronômico Andrei Cristian Ferreira, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Eduardo Camponogara
- Department of Automation and Systems Engineering, Federal University of Santa Catarina (UFSC), Engenheiro Agronômico Andrei Cristian Ferreira, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Viviana Cocco Mariani
- Electrical Engineering Graduate Program (PPGEE), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil; Mechanical Engineering Graduate Program (PGMec), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil
| | - Leandro Dos Santos Coelho
- Electrical Engineering Graduate Program (PPGEE), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil
| |
Collapse
|
20
|
Siuly S, Khare SK, Kabir E, Sadiq MT, Wang H. An efficient Parkinson's disease detection framework: Leveraging time-frequency representation and AlexNet convolutional neural network. Comput Biol Med 2024; 174:108462. [PMID: 38599069 DOI: 10.1016/j.compbiomed.2024.108462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/07/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder affecting the quality of life of over 10 million individuals worldwide. Early diagnosis is crucial for timely intervention and better patient outcomes. Electroencephalogram (EEG) signals are commonly used for early PD diagnosis due to their potential in monitoring disease progression. But traditional EEG-based methods lack exploration of brain regions that provide essential information about PD, and their performance falls short for real-time applications. To address these limitations, this study proposes a novel approach using a Time-Frequency Representation (TFR) based AlexNet Convolutional Neural Network (CNN) model to explore EEG channel-based analysis and identify critical brain regions efficiently diagnosing PD from EEG data. The Wavelet Scattering Transform (WST) is employed to capture distinct temporal and spectral characteristics, while AlexNet CNN is utilized to detect complex spatial patterns at different scales, accurately identifying intricate EEG patterns associated with PD. The experiment results on two real-time EEG PD datasets: San Diego dataset and the Iowa dataset demonstrate that frontal and central brain regions, including AF4 and AFz electrodes, contribute significantly to providing more representative features compared to other regions for PD detection. The proposed architecture achieves an impressive accuracy of 99.84% for the San Diego dataset and 95.79% for the Iowa dataset, outperforming existing EEG-based PD detection methods. The findings of this research will assist to create an essential technology for efficient PD diagnosis, enhancing patient care and quality of life.
Collapse
Affiliation(s)
- Siuly Siuly
- Institute for Sustainable Industries & Liveable Cities, Victoria University, Melbourne, Australia; Centre for Health Research, University of Southern Queensland, Toowoomba, Australia.
| | - Smith K Khare
- Mærsk Mc-Kinney Møller Institute, Faculty of Engineering, University of Southern Denmark, Denmark
| | - Enamul Kabir
- School of Mathematics, Physics and Computing, University of Southern Queensland, Toowoomba, Australia
| | - Muhammad Tariq Sadiq
- School of Computer Science and Electronic Engineering, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, United Kingdom
| | - Hua Wang
- Institute for Sustainable Industries & Liveable Cities, Victoria University, Melbourne, Australia
| |
Collapse
|
21
|
Nada H, Kim S, Lee K. PT-Finder: A multi-modal neural network approach to target identification. Comput Biol Med 2024; 174:108444. [PMID: 38636325 DOI: 10.1016/j.compbiomed.2024.108444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/20/2024]
Abstract
Efficient target identification for bioactive compounds, including novel synthetic analogs, is crucial for accelerating the drug discovery pipeline. However, the process of target identification presents significant challenges and is often expensive, which in turn can hinder the drug discovery efforts. To address these challenges machine learning applications have arisen as a promising approach for predicting the targets for novel chemical compounds. These methods allow the exploration of ligand-target interactions, uncovering of biochemical mechanisms, and the investigation of drug repurposing. Typically, the current target identification tools rely on assessing ligand structural similarities. Herein, a multi-modal neural network model was built using a library of proteins, their respective sequences, and active inhibitors. Subsequent validations showed the model to possess accuracy of 82 % and MPRAUC of 0.80. Leveraging the trained model, we developed PT-Finder (Protein Target Finder), a user-friendly offline application that is capable of predicting the target proteins for hundreds of compounds within a few seconds. This combination of offline operation, speed, and accuracy positions PT-Finder as a powerful tool to accelerate drug discovery workflows. PT-Finder and its source codes have been made freely accessible for download at https://github.com/PT-Finder/PT-Finder.
Collapse
Affiliation(s)
- Hossam Nada
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Sungdo Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Kyeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea.
| |
Collapse
|
22
|
Karampuri A, Kundur S, Perugu S. Exploratory drug discovery in breast cancer patients: A multimodal deep learning approach to identify novel drug candidates targeting RTK signaling. Comput Biol Med 2024; 174:108433. [PMID: 38642491 DOI: 10.1016/j.compbiomed.2024.108433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Breast cancer, a highly formidable and diverse malignancy predominantly affecting women globally, poses a significant threat due to its intricate genetic variability, rendering it challenging to diagnose accurately. Various therapies such as immunotherapy, radiotherapy, and diverse chemotherapy approaches like drug repurposing and combination therapy are widely used depending on cancer subtype and metastasis severity. Our study revolves around an innovative drug discovery strategy targeting potential drug candidates specific to RTK signalling, a prominently targeted receptor class in cancer. To accomplish this, we have developed a multimodal deep neural network (MM-DNN) based QSAR model integrating omics datasets to elucidate genomic, proteomic expression data, and drug responses, validated rigorously. The results showcase an R2 value of 0.917 and an RMSE value of 0.312, affirming the model's commendable predictive capabilities. Structural analogs of drug molecules specific to RTK signalling were sourced from the PubChem database, followed by meticulous screening to eliminate dissimilar compounds. Leveraging the MM-DNN-based QSAR model, we predicted the biological activity of these molecules, subsequently clustering them into three distinct groups. Feature importance analysis was performed. Consequently, we successfully identified prime drug candidates tailored for each potential downstream regulatory protein within the RTK signalling pathway. This method makes the early stages of drug development faster by removing inactive compounds, providing a hopeful path in combating breast cancer.
Collapse
Affiliation(s)
- Anush Karampuri
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India
| | - Sunitha Kundur
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India
| | - Shyam Perugu
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India.
| |
Collapse
|
23
|
Zhang H, Liu X, Cheng W, Wang T, Chen Y. Prediction of drug-target binding affinity based on deep learning models. Comput Biol Med 2024; 174:108435. [PMID: 38608327 DOI: 10.1016/j.compbiomed.2024.108435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
The prediction of drug-target binding affinity (DTA) plays an important role in drug discovery. Computerized virtual screening techniques have been used for DTA prediction, greatly reducing the time and economic costs of drug discovery. However, these techniques have not succeeded in reversing the low success rate of new drug development. In recent years, the continuous development of deep learning (DL) technology has brought new opportunities for drug discovery through the DTA prediction. This shift has moved the prediction of DTA from traditional machine learning methods to DL. The DL frameworks used for DTA prediction include convolutional neural networks (CNN), graph convolutional neural networks (GCN), and recurrent neural networks (RNN), and reinforcement learning (RL), among others. This review article summarizes the available literature on DTA prediction using DL models, including DTA quantification metrics and datasets, and DL algorithms used for DTA prediction (including input representation of models, neural network frameworks, valuation indicators, and model interpretability). In addition, the opportunities, challenges, and prospects of the application of DL frameworks for DTA prediction in the field of drug discovery are discussed.
Collapse
Affiliation(s)
- Hao Zhang
- College of Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoqian Liu
- College of Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wenya Cheng
- College of Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tianshi Wang
- College of Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yuanyuan Chen
- College of Science, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
24
|
D'Antona S, Porro D, Gallivanone F, Bertoli G. Characterization of cell cycle, inflammation, and oxidative stress signaling role in non-communicable diseases: Insights into genetic variants, microRNAs and pathways. Comput Biol Med 2024; 174:108346. [PMID: 38581999 DOI: 10.1016/j.compbiomed.2024.108346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/16/2024] [Accepted: 03/17/2024] [Indexed: 04/08/2024]
Abstract
Non-Communicable Diseases (NCDs) significantly impact global health, contributing to over 70% of premature deaths, as reported by the World Health Organization (WHO). These diseases have complex and multifactorial origins, involving genetic, epigenetic, environmental and lifestyle factors. While Genome-Wide Association Study (GWAS) is widely recognized as a valuable tool for identifying variants associated with complex phenotypes; the multifactorial nature of NCDs necessitates a more comprehensive exploration, encompassing not only the genetic but also the epigenetic aspect. For this purpose, we employed a bioinformatics-multiomics approach to examine the genetic and epigenetic characteristics of NCDs (i.e. colorectal cancer, coronary atherosclerosis, squamous cell lung cancer, psoriasis, type 2 diabetes, and multiple sclerosis), aiming to identify novel biomarkers for diagnosis and prognosis. Leveraging GWAS summary statistics, we pinpointed Single Nucleotide Polymorphisms (SNPs) independently associated with each NCD. Subsequently, we identified genes linked to cell cycle, inflammation and oxidative stress mechanisms, revealing shared genes across multiple diseases, suggesting common functional pathways. From an epigenetic perspective, we identified microRNAs (miRNAs) with regulatory functions targeting these genes of interest. Our findings underscore critical genetic pathways implicated in these diseases. In colorectal cancer, the dysregulation of the "Cytokine Signaling in Immune System" pathway, involving LAMA5 and SMAD7, regulated by Hsa-miR-21-5p, Hsa-miR-103a-3p, and Hsa-miR-195-5p, emerged as pivotal. In coronary atherosclerosis, the pathway associated with "binding of TCF/LEF:CTNNB1 to target gene promoters" displayed noteworthy implications, with the MYC factor controlled by Hsa-miR-16-5p as a potential regulatory factor. Squamous cell lung carcinoma analysis revealed significant pathways such as "PTK6 promotes HIF1A stabilization," regulated by Hsa-let-7b-5p. In psoriasis, the "Endosomal/Vacuolar pathway," involving HLA-C and Hsa-miR-148a-3p and Hsa-miR-148b-3p, was identified as crucial. Type 2 Diabetes implicated the "Regulation of TP53 Expression" pathway, controlled by Hsa-miR-106a-5p and Hsa-miR-106b-5p. In conclusion, our study elucidates the genetic framework and molecular mechanisms underlying NCDs, offering crucial insights into potential genetic/epigenetic biomarkers for diagnosis and prognosis. The specificity of pathways and related miRNAs in different pathologies highlights promising candidates for further clinical validation, with the potential to advance personalized treatments and alleviate the global burden of NCDs.
Collapse
Affiliation(s)
- Salvatore D'Antona
- Institute of Bioimaging and Molecular Physiology, National Research Council, Via F.lli Cervi 93, 20054, Milan, Italy
| | - Danilo Porro
- Institute of Bioimaging and Molecular Physiology, National Research Council, Via F.lli Cervi 93, 20054, Milan, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Francesca Gallivanone
- Institute of Bioimaging and Molecular Physiology, National Research Council, Via F.lli Cervi 93, 20054, Milan, Italy
| | - Gloria Bertoli
- Institute of Bioimaging and Molecular Physiology, National Research Council, Via F.lli Cervi 93, 20054, Milan, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy.
| |
Collapse
|
25
|
Chang C, Shi W, Wang Y, Zhang Z, Huang X, Jiao Y. The path from task-specific to general purpose artificial intelligence for medical diagnostics: A bibliometric analysis. Comput Biol Med 2024; 172:108258. [PMID: 38467093 DOI: 10.1016/j.compbiomed.2024.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/08/2024] [Accepted: 03/06/2024] [Indexed: 03/13/2024]
Abstract
Artificial intelligence (AI) has revolutionized many fields, and its potential in healthcare has been increasingly recognized. Based on diverse data sources such as imaging, laboratory tests, medical records, and electrophysiological data, diagnostic AI has witnessed rapid development in recent years. A comprehensive understanding of the development status, contributing factors, and their relationships in the application of AI to medical diagnostics is essential to further promote its use in clinical practice. In this study, we conducted a bibliometric analysis to explore the evolution of task-specific to general-purpose AI for medical diagnostics. We used the Web of Science database to search for relevant articles published between 2010 and 2023, and applied VOSviewer, the R package Bibliometrix, and CiteSpace to analyze collaborative networks and keywords. Our analysis revealed that the field of AI in medical diagnostics has experienced rapid growth in recent years, with a focus on tasks such as image analysis, disease prediction, and decision support. Collaborative networks were observed among researchers and institutions, indicating a trend of global cooperation in this field. Additionally, we identified several key factors contributing to the development of AI in medical diagnostics, including data quality, algorithm design, and computational power. Challenges to progress in the field include model explainability, robustness, and equality, which will require multi-stakeholder, interdisciplinary collaboration to tackle. Our study provides a holistic understanding of the path from task-specific, mono-modal AI toward general-purpose, multimodal AI for medical diagnostics. With the continuous improvement of AI technology and the accumulation of medical data, we believe that AI will play a greater role in medical diagnostics in the future.
Collapse
Affiliation(s)
- Chuheng Chang
- Department of General Practice (General Internal Medicine), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Wen Shi
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Youyang Wang
- Department of General Practice (General Internal Medicine), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Zhan Zhang
- Department of Computer Science and Technology, Tsinghua University, Beijing, China.
| | - Xiaoming Huang
- Department of General Practice (General Internal Medicine), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yang Jiao
- Department of General Practice (General Internal Medicine), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
26
|
Zhou Y, Chen Z, Yang M, Chen F, Yin J, Zhang Y, Zhou X, Sun X, Ni Z, Chen L, Lv Q, Zhu F, Liu S. FERREG: ferroptosis-based regulation of disease occurrence, progression and therapeutic response. Brief Bioinform 2024; 25:bbae223. [PMID: 38742521 PMCID: PMC11091744 DOI: 10.1093/bib/bbae223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 04/21/2024] [Indexed: 05/16/2024] Open
Abstract
Ferroptosis is a non-apoptotic, iron-dependent regulatory form of cell death characterized by the accumulation of intracellular reactive oxygen species. In recent years, a large and growing body of literature has investigated ferroptosis. Since ferroptosis is associated with various physiological activities and regulated by a variety of cellular metabolism and mitochondrial activity, ferroptosis has been closely related to the occurrence and development of many diseases, including cancer, aging, neurodegenerative diseases, ischemia-reperfusion injury and other pathological cell death. The regulation of ferroptosis mainly focuses on three pathways: system Xc-/GPX4 axis, lipid peroxidation and iron metabolism. The genes involved in these processes were divided into driver, suppressor and marker. Importantly, small molecules or drugs that mediate the expression of these genes are often good treatments in the clinic. Herein, a newly developed database, named 'FERREG', is documented to (i) providing the data of ferroptosis-related regulation of diseases occurrence, progression and drug response; (ii) explicitly describing the molecular mechanisms underlying each regulation; and (iii) fully referencing the collected data by cross-linking them to available databases. Collectively, FERREG contains 51 targets, 718 regulators, 445 ferroptosis-related drugs and 158 ferroptosis-related disease responses. FERREG can be accessed at https://idrblab.org/ferreg/.
Collapse
Affiliation(s)
- Yuan Zhou
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mengjie Yang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Fengyun Chen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jiayi Yin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xuheng Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Lu Chen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Qun Lv
- Department of Respiratory, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 311121, 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
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| |
Collapse
|
27
|
Zheng T, Zheng Z, Zhou H, Guo Y, Li S. The multifaceted roles of COL4A4 in lung adenocarcinoma: An integrated bioinformatics and experimental study. Comput Biol Med 2024; 170:107896. [PMID: 38217972 DOI: 10.1016/j.compbiomed.2023.107896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/03/2023] [Accepted: 12/23/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Abnormal expression of collagen IV subunits has been reported in cancers, but the significance is not clear. No study has reported the significance of COL4A4 in lung adenocarcinoma (LUAD). METHODS COL4A4 expression data, single-cell sequencing data and clinical data were downloaded from public databases. A range of bioinformatics and experimental methods were adopted to analyze the association of COL4A4 expression with clinical parameters, tumor microenvironment (TME), drug resistance and immunotherapy response, and to investigate the roles and underlying mechanism of COL4A4 in LUAD. RESULTS COL4A4 is differentially expressed in most of cancers analyzed, being associated with prognosis, tumor stemness, immune checkpoint gene expression and TME parameters. In LUAD, COL4A4 expression is down-regulated and associated with various TME parameters, response to immunotherapy and drug resistance. LUAD patients with lower COL4A4 have worse prognosis. Knockdown of COL4A4 significantly inhibited the expression of cell-cycle associated genes, and the expression and activation of signaling pathways including JAK/STAT3, p38, and ERK pathways, and induced quiescence in LUAD cells. Besides, it significantly induced the expression of a range of bioactive molecule genes that have been shown to have critical roles in TME remodeling and immune regulation. CONCLUSIONS COL4A4 is implicated in the pathogenesis of cancers including LUAD. Its function may be multifaceted. It can modulate the activity of LUAD cells, TME remodeling and tumor stemness, thus affecting the pathological process of LUAD. COL4A4 may be a prognostic molecular marker and a potential therapeutic target.
Collapse
Affiliation(s)
- Tiaozhan Zheng
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region, 530021, PR China
| | - Zhiwen Zheng
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region, 530021, PR China
| | - Hanxi Zhou
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang Province, PR China
| | - Yiqing Guo
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang Province, PR China
| | - Shikang Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region, 530021, PR China.
| |
Collapse
|
28
|
Chen B, Pan Z, Mou M, Zhou Y, Fu W. Is fragment-based graph a better graph-based molecular representation for drug design? A comparison study of graph-based models. Comput Biol Med 2024; 169:107811. [PMID: 38168647 DOI: 10.1016/j.compbiomed.2023.107811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/23/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024]
Abstract
Graph Neural Networks (GNNs) have gained significant traction in various sectors of AI-driven drug design. Over recent years, the integration of fragmentation concepts into GNNs has emerged as a potent strategy to augment the efficacy of molecular generative models. Nonetheless, challenges such as symmetry breaking and potential misrepresentation of intricate cycles and undefined functional groups raise questions about the superiority of fragment-based graph representation over traditional methods. In our research, we undertook a rigorous evaluation, contrasting the predictive prowess of eight models-developed using deep learning algorithms-across 12 benchmark datasets that span a range of properties. These models encompass established methods like GCN, AttentiveFP, and D-MPNN, as well as innovative fragment-based representation techniques. Our results indicate that fragment-based methodologies, notably PharmHGT, significantly improve model performance and interpretability, particularly in scenarios characterized by limited data availability. However, in situations with extensive training, fragment-based molecular graph representations may not necessarily eclipse traditional methods. In summation, we posit that the integration of fragmentation, as an avant-garde technique in drug design, harbors considerable promise for the future of AI-enhanced drug design.
Collapse
Affiliation(s)
- Baiyu Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 202103, China
| | - Ziqi Pan
- 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
| | - Yuan Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 202103, China.
| |
Collapse
|
29
|
Amahong K, Zhang W, Liu Y, Li T, Huang S, Han L, Tao L, Zhu F. RVvictor: Virus RNA-directed molecular interactions for RNA virus infection. Comput Biol Med 2024; 169:107886. [PMID: 38157777 DOI: 10.1016/j.compbiomed.2023.107886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
RNA viruses are major human pathogens that cause seasonal epidemics and occasional pandemic outbreaks. Due to the nature of their RNA genomes, it is anticipated that virus's RNA interacts with host protein (INTPRO), messenger RNA (INTmRNA), and non-coding RNA (INTncRNA) to perform their particular functions during their transcription and replication. In other words, thus, it is urgently needed to have such valuable data on virus RNA-directed molecular interactions (especially INTPROs), which are highly anticipated to attract broad research interests in the fields of RNA virus translation and replication. In this study, a new database was constructed to describe the virus RNA-directed interaction (INTPRO, INTmRNA, INTncRNA) for RNA virus (RVvictor). This database is unique in a) unambiguously characterizing the interactions between viruses RNAs and host proteins, b) providing, for the first time, the most systematic RNA-directed interaction data resources in providing clues to understand the molecular mechanisms of RNA viruses' translation, and replication, and c) in RVvictor, comprehensive enrichment analysis is conducted for each virus RNA based on its associated target genes/proteins, and the enrichment results were explicitly illustrated using various graphs. We found significant enrichment of a suite of pathways related to infection, translation, and replication, e.g., HIV infection, coronavirus disease, regulation of viral genome replication, and so on. Due to the devastating and persistent threat posed by the RNA virus, RVvictor constructed, for the first time, a possible network of cross-talk in RNA-directed interaction, which may ultimately explain the pathogenicity of RNA virus infection. The knowledge base might help develop new anti-viral therapeutic targets in the future. It's now free and publicly accessible at: https://idrblab.org/rvvictor/.
Collapse
Affiliation(s)
- Kuerbannisha Amahong
- 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
| | - Wei Zhang
- 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
| | - Yuhong Liu
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Teng Li
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Shijie Huang
- 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
| | - Lianyi Han
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, 315211, China.
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, 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
|