Artificial Intelligence (AI) in Biomedicine

A special issue of Biomolecules (ISSN 2218-273X). This special issue belongs to the section "Bioinformatics and Systems Biology".

Deadline for manuscript submissions: 30 September 2024 | Viewed by 13216

Special Issue Editor


E-Mail
Guest Editor
Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 106, Taiwan
Interests: artificial intelligence; bioinformatics; biomedical and healthcare informatics; genomics; medical imaging; proteomics; radiomics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Recently, artificial intelligence (AI) algorithms have shown promising results and advantages in processing various aspects of data. With the assistance of fast-improving AI algorithms (from machine learning, deep learning, natural language processing, etc.), we can use highly efficient data-mining tools to handle a huge body of biomedicine databases. AI in biomedicine includes both basic (i.e., biological and physiological principles) as well as clinical research with information of many biomedical disciplines and areas of speciality. Through these applications, AI expects to help transform the world of medicine by training models that predict how the phenotype and genotype are defined and enables the exploration of biomedical diagnosis and therapies.

This Special Issue aims to provide a place covering the applications of AI to different aspects of biomedicine. Research areas may include (but are not limited to) the following:

  • Applications of AI in molecular biology and biochemistry
  • Applications of AI in bioinformatics and system biology
  • Applications of AI in genomics and genetics
  • Applications of AI in drug discovery and development
  • Applications of AI in biomedical imaging
  • Applications of AI in disease diagnosis and prognosis
  • Applications of AI in clinical decision support systems
  • Applications of AI in biomedical and health informatics
  • Big data analysis in biomedicine
  • Natural language processing in biomedical text mining

Dr. Le Nguyen Quoc Khanh
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomolecules is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

19 pages, 1770 KiB  
Article
SynerGNet: A Graph Neural Network Model to Predict Anticancer Drug Synergy
by Mengmeng Liu, Gopal Srivastava, J. Ramanujam and Michal Brylinski
Biomolecules 2024, 14(3), 253; https://doi.org/10.3390/biom14030253 - 21 Feb 2024
Viewed by 1039
Abstract
Drug combination therapy shows promise in cancer treatment by addressing drug resistance, reducing toxicity, and enhancing therapeutic efficacy. However, the intricate and dynamic nature of biological systems makes identifying potential synergistic drugs a costly and time-consuming endeavor. To facilitate the development of combination [...] Read more.
Drug combination therapy shows promise in cancer treatment by addressing drug resistance, reducing toxicity, and enhancing therapeutic efficacy. However, the intricate and dynamic nature of biological systems makes identifying potential synergistic drugs a costly and time-consuming endeavor. To facilitate the development of combination therapy, techniques employing artificial intelligence have emerged as a transformative solution, providing a sophisticated avenue for advancing existing therapeutic approaches. In this study, we developed SynerGNet, a graph neural network model designed to accurately predict the synergistic effect of drug pairs against cancer cell lines. SynerGNet utilizes cancer-specific featured graphs created by integrating heterogeneous biological features into the human protein–protein interaction network, followed by a reduction process to enhance topological diversity. Leveraging synergy data provided by AZ-DREAM Challenges, the model yields a balanced accuracy of 0.68, significantly outperforming traditional machine learning. Encouragingly, augmenting the training data with carefully constructed synthetic instances improved the balanced accuracy of SynerGNet to 0.73. Finally, the results of an independent validation conducted against DrugCombDB demonstrated that it exhibits a strong performance when applied to unseen data. SynerGNet shows a great potential in detecting drug synergy, positioning itself as a valuable tool that could contribute to the advancement of combination therapy for cancer treatment. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

12 pages, 1923 KiB  
Article
Combining Low-Dose Computer-Tomography-Based Radiomics and Serum Metabolomics for Diagnosis of Malignant Nodules in Participants of Lung Cancer Screening Studies
by Joanna Zyla, Michal Marczyk, Wojciech Prazuch, Magdalena Sitkiewicz, Agata Durawa, Malgorzata Jelitto, Katarzyna Dziadziuszko, Karol Jelonek, Agata Kurczyk, Edyta Szurowska, Witold Rzyman, Piotr Widłak and Joanna Polanska
Biomolecules 2024, 14(1), 44; https://doi.org/10.3390/biom14010044 - 28 Dec 2023
Viewed by 1082
Abstract
Radiomics is an emerging approach to support the diagnosis of pulmonary nodules detected via low-dose computed tomography lung cancer screening. Serum metabolome is a promising source of auxiliary biomarkers that could help enhance the precision of lung cancer diagnosis in CT-based screening. Thus, [...] Read more.
Radiomics is an emerging approach to support the diagnosis of pulmonary nodules detected via low-dose computed tomography lung cancer screening. Serum metabolome is a promising source of auxiliary biomarkers that could help enhance the precision of lung cancer diagnosis in CT-based screening. Thus, we aimed to verify whether the combination of these two techniques, which provides local/morphological and systemic/molecular features of disease at the same time, increases the performance of lung cancer classification models. The collected cohort consists of 1086 patients with radiomic and 246 patients with serum metabolomic evaluations. Different machine learning techniques, i.e., random forest and logistic regression were applied for each omics. Next, model predictions were combined with various integration methods to create a final model. The best single omics models were characterized by an AUC of 83% in radiomics and 60% in serum metabolomics. The model integration only slightly increased the performance of the combined model (AUC equal to 85%), which was not statistically significant. We concluded that radiomics itself has a good ability to discriminate lung cancer from benign lesions. However, additional research is needed to test whether its combination with other molecular assessments would further improve the diagnosis of screening-detected lung nodules. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

17 pages, 4278 KiB  
Article
ConF: A Deep Learning Model Based on BiLSTM, CNN, and Cross Multi-Head Attention Mechanism for Noncoding RNA Family Prediction
by Shoryu Teragawa and Lei Wang
Biomolecules 2023, 13(11), 1643; https://doi.org/10.3390/biom13111643 - 13 Nov 2023
Viewed by 959
Abstract
This paper presents ConF, a novel deep learning model designed for accurate and efficient prediction of noncoding RNA families. NcRNAs are essential functional RNA molecules involved in various cellular processes, including replication, transcription, and gene expression. Identifying ncRNA families is crucial for comprehensive [...] Read more.
This paper presents ConF, a novel deep learning model designed for accurate and efficient prediction of noncoding RNA families. NcRNAs are essential functional RNA molecules involved in various cellular processes, including replication, transcription, and gene expression. Identifying ncRNA families is crucial for comprehensive RNA research, as ncRNAs within the same family often exhibit similar functionalities. Traditional experimental methods for identifying ncRNA families are time-consuming and labor-intensive. Computational approaches relying on annotated secondary structure data face limitations in handling complex structures like pseudoknots and have restricted applicability, resulting in suboptimal prediction performance. To overcome these challenges, ConF integrates mainstream techniques such as residual networks with dilated convolutions and cross multi-head attention mechanisms. By employing a combination of dual-layer convolutional networks and BiLSTM, ConF effectively captures intricate features embedded within RNA sequences. This feature extraction process leads to significantly improved prediction accuracy compared to existing methods. Experimental evaluations conducted using a single, publicly available dataset and applying ten-fold cross-validation demonstrate the superiority of ConF in terms of accuracy, sensitivity, and other performance metrics. Overall, ConF represents a promising solution for accurate and efficient ncRNA family prediction, addressing the limitations of traditional experimental and computational methods. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

18 pages, 2479 KiB  
Article
Transformer-Based Multi-Modal Data Fusion Method for COPD Classification and Physiological and Biochemical Indicators Identification
by Weidong Xie, Yushan Fang, Guicheng Yang, Kun Yu and Wei Li
Biomolecules 2023, 13(9), 1391; https://doi.org/10.3390/biom13091391 - 15 Sep 2023
Cited by 1 | Viewed by 1225
Abstract
As the number of modalities in biomedical data continues to increase, the significance of multi-modal data becomes evident in capturing complex relationships between biological processes, thereby complementing disease classification. However, the current multi-modal fusion methods for biomedical data require more effective exploitation of [...] Read more.
As the number of modalities in biomedical data continues to increase, the significance of multi-modal data becomes evident in capturing complex relationships between biological processes, thereby complementing disease classification. However, the current multi-modal fusion methods for biomedical data require more effective exploitation of intra- and inter-modal interactions, and the application of powerful fusion methods to biomedical data is relatively rare. In this paper, we propose a novel multi-modal data fusion method that addresses these limitations. Our proposed method utilizes a graph neural network and a 3D convolutional network to identify intra-modal relationships. By doing so, we can extract meaningful features from each modality, preserving crucial information. To fuse information from different modalities, we employ the Low-rank Multi-modal Fusion method, which effectively integrates multiple modalities while reducing noise and redundancy. Additionally, our method incorporates the Cross-modal Transformer to automatically learn relationships between different modalities, facilitating enhanced information exchange and representation. We validate the effectiveness of our proposed method using lung CT imaging data and physiological and biochemical data obtained from patients diagnosed with Chronic Obstructive Pulmonary Disease (COPD). Our method demonstrates superior performance compared to various fusion methods and their variants in terms of disease classification accuracy. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

15 pages, 3257 KiB  
Article
Molecular Property Prediction by Combining LSTM and GAT
by Lei Xu, Shourun Pan, Leiming Xia and Zhen Li
Biomolecules 2023, 13(3), 503; https://doi.org/10.3390/biom13030503 - 09 Mar 2023
Cited by 6 | Viewed by 2454
Abstract
Molecular property prediction is an important direction in computer-aided drug design. In this paper, to fully explore the information from SMILE stings and graph data of molecules, we combined the SALSTM and GAT methods in order to mine the feature information of molecules [...] Read more.
Molecular property prediction is an important direction in computer-aided drug design. In this paper, to fully explore the information from SMILE stings and graph data of molecules, we combined the SALSTM and GAT methods in order to mine the feature information of molecules from sequences and graphs. The embedding atoms are obtained through SALSTM, firstly using SMILES strings, and they are combined with graph node features and fed into the GAT to extract the global molecular representation. At the same time, data augmentation is added to enlarge the training dataset and improve the performance of the model. Finally, to enhance the interpretability of the model, the attention layers of both models are fused together to highlight the key atoms. Comparison with other graph-based and sequence-based methods, for multiple datasets, shows that our method can achieve high prediction accuracy with good generalizability. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

14 pages, 2300 KiB  
Article
G4Beacon: An In Vivo G4 Prediction Method Using Chromatin and Sequence Information
by Zhuofan Zhang, Rongxin Zhang, Ke Xiao and Xiao Sun
Biomolecules 2023, 13(2), 292; https://doi.org/10.3390/biom13020292 - 03 Feb 2023
Cited by 2 | Viewed by 1770
Abstract
G-quadruplex (G4) structures are critical epigenetic regulatory elements, which usually form in guanine-rich regions in DNA. However, predicting the formation of G4 structures within living cells remains a challenge. Here, we present an ultra-robust machine learning method, G4Beacon, which utilizes the Gradient-Boosting Decision [...] Read more.
G-quadruplex (G4) structures are critical epigenetic regulatory elements, which usually form in guanine-rich regions in DNA. However, predicting the formation of G4 structures within living cells remains a challenge. Here, we present an ultra-robust machine learning method, G4Beacon, which utilizes the Gradient-Boosting Decision Tree (GBDT) algorithm, coupled with the ATAC-seq data and the surrounding sequences of in vitro G4s, to accurately predict the formation ability of these in vitro G4s in different cell types. As a result, our model achieved excellent performance even when the test set was extremely skewed. Besides this, G4Beacon can also identify the in vivo G4s of other cell lines precisely with the model built on a special cell line, regardless of the experimental techniques or platforms. Altogether, G4Beacon is an accurate, reliable, and easy-to-use method for the prediction of in vivo G4s of various cell lines. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

14 pages, 1814 KiB  
Article
A Machine Learning Approach for Recommending Herbal Formulae with Enhanced Interpretability and Applicability
by Won-Yung Lee, Youngseop Lee, Siwoo Lee, Young Woo Kim and Ji-Hwan Kim
Biomolecules 2022, 12(11), 1604; https://doi.org/10.3390/biom12111604 - 31 Oct 2022
Cited by 1 | Viewed by 1556
Abstract
Herbal formulae (HFs) are representative interventions in Korean medicine (KM) for the prevention and treatment of various diseases. Here, we proposed a machine learning-based approach for HF recommendation with enhanced interpretability and applicability. A dataset consisting of clinical symptoms, Sasang constitution (SC) types, [...] Read more.
Herbal formulae (HFs) are representative interventions in Korean medicine (KM) for the prevention and treatment of various diseases. Here, we proposed a machine learning-based approach for HF recommendation with enhanced interpretability and applicability. A dataset consisting of clinical symptoms, Sasang constitution (SC) types, and prescribed HFs was derived from a multicenter study. Case studies published over 10 years were collected and curated by experts. Various classifiers, oversampling methods, and data imputation techniques were comprehensively considered. The local interpretable model-agnostic explanation (LIME) technique was applied to identify the clinical symptoms that led to the recommendation of specific HFs. We found that the cascaded deep forest (CDF) model with data imputation and oversampling yielded the best performance on the training set and holdout test set. Our model also achieved top-1 and top-3 accuracies of 0.35 and 0.89, respectively, on case study datasets in which clinical symptoms were only partially recorded. We performed an expert evaluation on the reliability of interpretation results using case studies and achieved a score close to normal. Taken together, our model will contribute to the modernization of KM and the identification of an HF selection process through the development of a practically useful HF recommendation model. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

Review

Jump to: Research

27 pages, 2552 KiB  
Review
A Review for Artificial Intelligence Based Protein Subcellular Localization
by Hanyu Xiao, Yijin Zou, Jieqiong Wang and Shibiao Wan
Biomolecules 2024, 14(4), 409; https://doi.org/10.3390/biom14040409 - 27 Mar 2024
Viewed by 709
Abstract
Proteins need to be located in appropriate spatiotemporal contexts to carry out their diverse biological functions. Mislocalized proteins may lead to a broad range of diseases, such as cancer and Alzheimer’s disease. Knowing where a target protein resides within a cell will give [...] Read more.
Proteins need to be located in appropriate spatiotemporal contexts to carry out their diverse biological functions. Mislocalized proteins may lead to a broad range of diseases, such as cancer and Alzheimer’s disease. Knowing where a target protein resides within a cell will give insights into tailored drug design for a disease. As the gold validation standard, the conventional wet lab uses fluorescent microscopy imaging, immunoelectron microscopy, and fluorescent biomarker tags for protein subcellular location identification. However, the booming era of proteomics and high-throughput sequencing generates tons of newly discovered proteins, making protein subcellular localization by wet-lab experiments a mission impossible. To tackle this concern, in the past decades, artificial intelligence (AI) and machine learning (ML), especially deep learning methods, have made significant progress in this research area. In this article, we review the latest advances in AI-based method development in three typical types of approaches, including sequence-based, knowledge-based, and image-based methods. We also elaborately discuss existing challenges and future directions in AI-based method development in this research field. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

Back to TopTop