Pub Date : 2024-11-20DOI: 10.1016/j.compbiomed.2024.109389
Sara Sweidan, S S Askar, Mohamed Abouhawwash, Elsayed Badr
Obesity is a chronic disease correlated with numerous risk factors that not only negatively affect all body functions but also increase the chances of developing chronic diseases and the associated morbidity and mortality rates. This study proposes a novel system that bridges the gap between healthcare providers and patients by offering both parties some tools for navigating the intricacies of dietary planning. In this system, machine learning techniques are used to determine the required calories before starting an obesity treatment. A hybrid precision model with minimal parameters is also developed to estimate the appropriate number of calories for losing weight and to formulate a healthy diet plan. A real dataset of 15 anthropometric measurements is analyzed using SVR, LR, and DTR regression models, and all the data are preprocessed before analysis to enhance model performance. Results show that the required calories can be estimated with a high correlation (R = 0.985) from independent measurements. The proposed model also calculates the healthy daily percentages of fats, proteins, and carbohydrates based on a knowledge base of medical rules and functions, thus facilitating the sequential treatment of obese patients. In sum, this study applies different models to design a practical, cost-effective approach for accurately determining the required calories and formulating valuable diet plans for obesity treatment and management.
{"title":"A hybrid healthy diet recommender system based on machine learning techniques.","authors":"Sara Sweidan, S S Askar, Mohamed Abouhawwash, Elsayed Badr","doi":"10.1016/j.compbiomed.2024.109389","DOIUrl":"https://doi.org/10.1016/j.compbiomed.2024.109389","url":null,"abstract":"<p><p>Obesity is a chronic disease correlated with numerous risk factors that not only negatively affect all body functions but also increase the chances of developing chronic diseases and the associated morbidity and mortality rates. This study proposes a novel system that bridges the gap between healthcare providers and patients by offering both parties some tools for navigating the intricacies of dietary planning. In this system, machine learning techniques are used to determine the required calories before starting an obesity treatment. A hybrid precision model with minimal parameters is also developed to estimate the appropriate number of calories for losing weight and to formulate a healthy diet plan. A real dataset of 15 anthropometric measurements is analyzed using SVR, LR, and DTR regression models, and all the data are preprocessed before analysis to enhance model performance. Results show that the required calories can be estimated with a high correlation (R = 0.985) from independent measurements. The proposed model also calculates the healthy daily percentages of fats, proteins, and carbohydrates based on a knowledge base of medical rules and functions, thus facilitating the sequential treatment of obese patients. In sum, this study applies different models to design a practical, cost-effective approach for accurately determining the required calories and formulating valuable diet plans for obesity treatment and management.</p>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"109389"},"PeriodicalIF":7.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-19DOI: 10.1016/j.compbiomed.2024.109397
Damaris Rivera-Asencios, Abraham Espinoza-Culupú, Sheyla Carmen-Sifuentes, Pablo Ramirez, Ruth García-de-la-Guarda
Carrion's disease, caused by the bacterium Bartonella bacilliformis, is a serious public health problem in Peru, Ecuador and Colombia. Currently there is no available vaccine against B. bacilliformis. While antibiotics are the standard treatment, resistant strains have been reported, and there is a potential spread of the vector that transmits the bacteria. This study aimed to design a multi-epitope vaccine candidate against the causative agent of Carrion's disease using immunoinformatics tools. Predictions of B-cell epitopes, as well as CD4+ and CD8+T cell epitopes, were performed from the entire proteome of B. bacilliformis KC583 using the most frequent alleles from Peru, Ecuador, Colombia, and worldwide. B-cell epitopes and T-cell nested epitopes from outer membrane and virulence-associated proteins were selected. Epitopes were filtered out based on promiscuity, non-allergenicity, conservation, non-homology and non-toxicity. Two vaccine constructs were assembled using linkers. The tertiary structure of the constructs was predicted, and their stability was evaluated through molecular dynamics simulations. The most stable construct was selected for molecular docking with the TLR4 receptor. This study proposes a vaccine construct evaluated in silico as a potential vaccine candidate against Bartonella bacilliformis.
由巴氏杆菌引起的卡里翁病是秘鲁、厄瓜多尔和哥伦比亚的一个严重公共卫生问题。目前还没有针对巴氏杆菌的疫苗。虽然抗生素是标准的治疗方法,但耐药菌株已有报道,而且传播细菌的病媒也有可能扩散。本研究旨在利用免疫信息学工具设计一种针对卡里昂氏病病原体的多表位候选疫苗。研究人员利用秘鲁、厄瓜多尔、哥伦比亚和全球最常见的等位基因,从杆菌 KC583 的整个蛋白质组中预测了 B 细胞表位以及 CD4+ 和 CD8+T 细胞表位。从外膜蛋白和毒力相关蛋白中筛选出了 B 细胞表位和 T 细胞嵌套表位。表位是根据杂合性、非致敏性、保护性、非同源性和无毒性筛选出来的。使用连接体组装了两个疫苗构建体。对构建体的三级结构进行了预测,并通过分子动力学模拟对其稳定性进行了评估。选择了最稳定的构建物与 TLR4 受体进行分子对接。本研究提出了一种疫苗构建体,并对其进行了硅学评估,认为它是一种潜在的巴氏杆菌候选疫苗。
{"title":"Design of a multi-epitope vaccine candidate against carrion disease by immunoinformatics approach.","authors":"Damaris Rivera-Asencios, Abraham Espinoza-Culupú, Sheyla Carmen-Sifuentes, Pablo Ramirez, Ruth García-de-la-Guarda","doi":"10.1016/j.compbiomed.2024.109397","DOIUrl":"https://doi.org/10.1016/j.compbiomed.2024.109397","url":null,"abstract":"<p><p>Carrion's disease, caused by the bacterium Bartonella bacilliformis, is a serious public health problem in Peru, Ecuador and Colombia. Currently there is no available vaccine against B. bacilliformis. While antibiotics are the standard treatment, resistant strains have been reported, and there is a potential spread of the vector that transmits the bacteria. This study aimed to design a multi-epitope vaccine candidate against the causative agent of Carrion's disease using immunoinformatics tools. Predictions of B-cell epitopes, as well as CD4<sup>+</sup> and CD8<sup>+</sup>T cell epitopes, were performed from the entire proteome of B. bacilliformis KC583 using the most frequent alleles from Peru, Ecuador, Colombia, and worldwide. B-cell epitopes and T-cell nested epitopes from outer membrane and virulence-associated proteins were selected. Epitopes were filtered out based on promiscuity, non-allergenicity, conservation, non-homology and non-toxicity. Two vaccine constructs were assembled using linkers. The tertiary structure of the constructs was predicted, and their stability was evaluated through molecular dynamics simulations. The most stable construct was selected for molecular docking with the TLR4 receptor. This study proposes a vaccine construct evaluated in silico as a potential vaccine candidate against Bartonella bacilliformis.</p>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"109397"},"PeriodicalIF":7.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-19DOI: 10.1016/j.compbiomed.2024.109415
Emerson Lucena da Silva, Felipe Pantoja Mesquita, Laine Celestino Pinto, Bruna Puty Silva Gomes, Edivaldo Herculano Correa de Oliveira, Rommel Mario Rodríguez Burbano, Maria Elisabete Amaral de Moares, Pedro Filho Noronha de Souza, Raquel Carvalho Montenegro
Gastric cancer (GC) is a common cancer worldwide. Therefore, searching for effective treatments is essential, and drug repositioning can be a promising strategy to find new potential drugs for GC therapy. For the first time, we sought to identify molecular alterations and validate new mechanisms related to Mebendazole (MBZ) treatment in GC cells through transcriptome analysis using microarray technology. Data revealed 1066 differentially expressed genes (DEGs), of which 345 (2.41 %) genes were upregulated, 721 (5.04 %) genes were downregulated, and 13,231 (92.54 %) genes remained unaltered after MBZ exposure. The overexpressed genes identified were CCL2, IL1A, and CDKN1A. In contrast, the H3C7, H3C11, and H1-5 were the top 3 underexpressed genes. Gene set enrichment analysis (GSEA) identified 8 pathways significantly overexpressed in the treated group (p < 0.05 and FDR<0.25). The validation of the expression of top desregulated genes by RT-qPCR confirmed the transcriptome results, where MBZ increased the CCL2, IL1A, and CDKN1A and reduced the H3C7, H3C11, and H1-5 transcript levels. Expression analysis in samples from TCGA databases correlated that the lower ILI1A and higher H3C11 and H1-5 gene expression are associated with decreased overall survival rates in patients with GC, indicating that MBZ treatment can improve the prognosis of patients. Thus, the data demonstrated that the drug MBZ alters the transcriptome of the AGP-01 lineage, mainly modulating the expression of histone proteins and inflammatory cytokines, indicating a possible epigenetic and immunological effect on tumor cells, these findings highlight new mechanisms of action related to MBZ treatment. Additional studies are still needed to better clarify the epigenetic and immune mechanism of MBZ in the therapy of GC.
{"title":"Transcriptome analysis displays new molecular insights into the mechanisms of action of Mebendazole in gastric cancer cells.","authors":"Emerson Lucena da Silva, Felipe Pantoja Mesquita, Laine Celestino Pinto, Bruna Puty Silva Gomes, Edivaldo Herculano Correa de Oliveira, Rommel Mario Rodríguez Burbano, Maria Elisabete Amaral de Moares, Pedro Filho Noronha de Souza, Raquel Carvalho Montenegro","doi":"10.1016/j.compbiomed.2024.109415","DOIUrl":"https://doi.org/10.1016/j.compbiomed.2024.109415","url":null,"abstract":"<p><p>Gastric cancer (GC) is a common cancer worldwide. Therefore, searching for effective treatments is essential, and drug repositioning can be a promising strategy to find new potential drugs for GC therapy. For the first time, we sought to identify molecular alterations and validate new mechanisms related to Mebendazole (MBZ) treatment in GC cells through transcriptome analysis using microarray technology. Data revealed 1066 differentially expressed genes (DEGs), of which 345 (2.41 %) genes were upregulated, 721 (5.04 %) genes were downregulated, and 13,231 (92.54 %) genes remained unaltered after MBZ exposure. The overexpressed genes identified were CCL2, IL1A, and CDKN1A. In contrast, the H3C7, H3C11, and H1-5 were the top 3 underexpressed genes. Gene set enrichment analysis (GSEA) identified 8 pathways significantly overexpressed in the treated group (p < 0.05 and FDR<0.25). The validation of the expression of top desregulated genes by RT-qPCR confirmed the transcriptome results, where MBZ increased the CCL2, IL1A, and CDKN1A and reduced the H3C7, H3C11, and H1-5 transcript levels. Expression analysis in samples from TCGA databases correlated that the lower ILI1A and higher H3C11 and H1-5 gene expression are associated with decreased overall survival rates in patients with GC, indicating that MBZ treatment can improve the prognosis of patients. Thus, the data demonstrated that the drug MBZ alters the transcriptome of the AGP-01 lineage, mainly modulating the expression of histone proteins and inflammatory cytokines, indicating a possible epigenetic and immunological effect on tumor cells, these findings highlight new mechanisms of action related to MBZ treatment. Additional studies are still needed to better clarify the epigenetic and immune mechanism of MBZ in the therapy of GC.</p>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"109415"},"PeriodicalIF":7.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681161","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-19DOI: 10.1016/j.compbiomed.2024.109432
Shahriar Mohammadi, Mohammad Ahmadi Livani
Breast mass segmentation plays a crucial role in early breast cancer detection and diagnosis, and while Convolutional Neural Networks (CNN) have been widely used for this task, their reliance on local receptive fields limits ability to capture long-range dependencies. Vision Transformers (ViTs), on the other hand, excel in this area by leveraging multi-head self-attention mechanisms to generate attention maps that dynamically gather global spatial information, significantly outperforming CNN-based architectures in various tasks. However, traditional transformer-based models come with challenges, including high computational complexity due to the self-attention mechanism and inefficiency in the static MLP fusion process. To overcome these issues, the Hybrid Transformer U-Net (HTU-net) model is proposed for breast mass segmentation in mammography. Channel and spatial enhanced self-attention mechanisms are integrated with convolutions layers in HTU-Net, creating a hybrid architecture that combines the strengths of both CNNs and ViTs. The introduction of a multiscale attention mechanism further improves the model's ability to fuse information from different resolutions, enhancing the decoder's capacity to reconstruct fine details in the segmented output. By using both local texture-based features and global contextual information, HTU-Net excels in capturing essential features, thus improving segmentation performance. The experimental results across multiple datasets, including CBIS-DDSM and INbreast, demonstrate that HTU-Net outperforms several state-of-the-art methods, achieving superior accuracy, dice similarity coefficient, and intersection over union. This work highlights the potential of hybrid architectures in advancing computer-aided diagnosis systems, particularly in improving segmentation quality and reliability for breast cancer detection.
乳房肿块分割在早期乳腺癌检测和诊断中起着至关重要的作用,虽然卷积神经网络(CNN)已被广泛应用于这项任务,但其对局部感受野的依赖限制了捕捉长距离依赖关系的能力。另一方面,视觉变换器(ViTs)在这一领域表现出色,它利用多头自我注意机制生成可动态收集全局空间信息的注意图,在各种任务中明显优于基于 CNN 的架构。然而,传统的基于变压器的模型也面临挑战,包括自注意机制导致的高计算复杂性和静态 MLP 融合过程的低效率。为了克服这些问题,我们提出了混合变换器 U-Net 模型(HTU-net),用于乳腺 X 射线摄影中的乳房肿块分割。HTU-Net 中的卷积层集成了通道和空间增强自注意机制,形成了一种混合架构,结合了 CNN 和 ViT 的优势。多尺度注意机制的引入进一步提高了模型融合不同分辨率信息的能力,增强了解码器在分割输出中重建精细细节的能力。通过同时使用基于纹理的局部特征和全局上下文信息,HTU-Net 在捕捉基本特征方面表现出色,从而提高了分割性能。包括 CBIS-DDSM 和 INbreast 在内的多个数据集的实验结果表明,HTU-Net 的表现优于几种最先进的方法,在准确性、骰子相似系数和相交优于联合方面都表现出色。这项工作凸显了混合架构在推进计算机辅助诊断系统方面的潜力,特别是在提高乳腺癌检测的分割质量和可靠性方面。
{"title":"Enhanced breast mass segmentation in mammograms using a hybrid transformer UNet model.","authors":"Shahriar Mohammadi, Mohammad Ahmadi Livani","doi":"10.1016/j.compbiomed.2024.109432","DOIUrl":"https://doi.org/10.1016/j.compbiomed.2024.109432","url":null,"abstract":"<p><p>Breast mass segmentation plays a crucial role in early breast cancer detection and diagnosis, and while Convolutional Neural Networks (CNN) have been widely used for this task, their reliance on local receptive fields limits ability to capture long-range dependencies. Vision Transformers (ViTs), on the other hand, excel in this area by leveraging multi-head self-attention mechanisms to generate attention maps that dynamically gather global spatial information, significantly outperforming CNN-based architectures in various tasks. However, traditional transformer-based models come with challenges, including high computational complexity due to the self-attention mechanism and inefficiency in the static MLP fusion process. To overcome these issues, the Hybrid Transformer U-Net (HTU-net) model is proposed for breast mass segmentation in mammography. Channel and spatial enhanced self-attention mechanisms are integrated with convolutions layers in HTU-Net, creating a hybrid architecture that combines the strengths of both CNNs and ViTs. The introduction of a multiscale attention mechanism further improves the model's ability to fuse information from different resolutions, enhancing the decoder's capacity to reconstruct fine details in the segmented output. By using both local texture-based features and global contextual information, HTU-Net excels in capturing essential features, thus improving segmentation performance. The experimental results across multiple datasets, including CBIS-DDSM and INbreast, demonstrate that HTU-Net outperforms several state-of-the-art methods, achieving superior accuracy, dice similarity coefficient, and intersection over union. This work highlights the potential of hybrid architectures in advancing computer-aided diagnosis systems, particularly in improving segmentation quality and reliability for breast cancer detection.</p>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"109432"},"PeriodicalIF":7.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681157","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Single-cell RNA sequencing (scRNA-seq) technology establishes a unique view for elucidating cellular heterogeneity in various biological systems. Yet the scRNA-seq data is compromised by a high dropout rate due to the technological limitation, and the substantial data loss poses computational challenges on subsequent analyses. This study introduces a novel graph clustering autoencoder (GCAE)-based imputation approach (GraCEImpute) to address the challenge of missing data in scRNA-seq data. Our comprehensive evaluation demonstrates that the GraCEImpute model outperforms existing approaches in accurately imputing dropout zeros within scRNA-seq data. The proposed GraCEImpute model also demonstrates the significantly enhanced quality of downstream scRNA-seq data analyses, including clustering, differential gene expression (DEG) analysis, and cell trajectory inference. These improvements underscore the GraCEImpute model's potential to facilitate a deeper understanding of cellular processes and heterogeneity through the scRNA-seq data analyses. The source code is released at https://www.healthinformaticslab.org/supp/.
{"title":"GraCEImpute: A novel graph clustering autoencoder approach for imputation of single-cell RNA-seq data.","authors":"Yueying Wang, Kewei Li, Ruochi Zhang, Yusi Fan, Lan Huang, Fengfeng Zhou","doi":"10.1016/j.compbiomed.2024.109400","DOIUrl":"https://doi.org/10.1016/j.compbiomed.2024.109400","url":null,"abstract":"<p><p>Single-cell RNA sequencing (scRNA-seq) technology establishes a unique view for elucidating cellular heterogeneity in various biological systems. Yet the scRNA-seq data is compromised by a high dropout rate due to the technological limitation, and the substantial data loss poses computational challenges on subsequent analyses. This study introduces a novel graph clustering autoencoder (GCAE)-based imputation approach (GraCEImpute) to address the challenge of missing data in scRNA-seq data. Our comprehensive evaluation demonstrates that the GraCEImpute model outperforms existing approaches in accurately imputing dropout zeros within scRNA-seq data. The proposed GraCEImpute model also demonstrates the significantly enhanced quality of downstream scRNA-seq data analyses, including clustering, differential gene expression (DEG) analysis, and cell trajectory inference. These improvements underscore the GraCEImpute model's potential to facilitate a deeper understanding of cellular processes and heterogeneity through the scRNA-seq data analyses. The source code is released at https://www.healthinformaticslab.org/supp/.</p>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"109400"},"PeriodicalIF":7.0,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674707","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-18DOI: 10.1016/j.compbiomed.2024.109308
Milad Habibi , Seda Aslan , Xiaolong Liu , Yue-Hin Loke , Axel Krieger , Narutoshi Hibino , Laura Olivieri , Mark Fuge
Cognitional heart disease is one of the leading causes of mortality among newborns. Tissue-engineered vascular grafts offer the potential to help treat cognitional heart disease through patient-specific vascular grafts. However, current methods often rely on non-personalized designs or involve significant human intervention. This paper presents a computational framework for the automatic shape optimization of patient-specific tissue-engineered vascular grafts for repairing the aortic arch, aimed at reducing the need for manual input and improving current treatment outcomes, which either use non-patient-specific geometry or require extensive human intervention to design the vascular graft. The paper’s core innovation lies in an automatic shape optimization pipeline that combines Bayesian optimization techniques with the open-source finite volume solver, OpenFOAM, and a novel graft deformation algorithm. Specifically, our framework begins with Laplacian mode computation and the approximation of a computationally low-cost Gaussian process surrogate model to capture the minimum weighted combination of inlet-outlet pressure drop (PD) and maximum wall shear stress (WSS). Bayesian Optimization then performs a limited number of OpenFOAM simulations to identify the optimal patient-specific shape.
We use imaging and flow data obtained from six patients diagnosed with cognitional heart disease to evaluate our approach. Our results showcase the potential of online training and hemodynamic surrogate model optimization for providing optimal graft shapes. These results show how our framework successfully reduces inlet-outlet PD and maximum WSS compared to pre-lofted models that include both the native geometry and human-designed grafts. Furthermore, we compare how the performance of each design optimized under steady-state simulation compares to that design’s performance under transient simulation, and to what extent the optimal design remains similar under both conditions. Our findings underscore that the automated designs achieve at least a 16% reduction in blood flow pressure drop in comparison to geometries optimized by humans.
{"title":"Automatic Laplacian-based shape optimization for patient-specific vascular grafts","authors":"Milad Habibi , Seda Aslan , Xiaolong Liu , Yue-Hin Loke , Axel Krieger , Narutoshi Hibino , Laura Olivieri , Mark Fuge","doi":"10.1016/j.compbiomed.2024.109308","DOIUrl":"10.1016/j.compbiomed.2024.109308","url":null,"abstract":"<div><div>Cognitional heart disease is one of the leading causes of mortality among newborns. Tissue-engineered vascular grafts offer the potential to help treat cognitional heart disease through patient-specific vascular grafts. However, current methods often rely on non-personalized designs or involve significant human intervention. This paper presents a computational framework for the automatic shape optimization of patient-specific tissue-engineered vascular grafts for repairing the aortic arch, aimed at reducing the need for manual input and improving current treatment outcomes, which either use non-patient-specific geometry or require extensive human intervention to design the vascular graft. The paper’s core innovation lies in an automatic shape optimization pipeline that combines Bayesian optimization techniques with the open-source finite volume solver, OpenFOAM, and a novel graft deformation algorithm. Specifically, our framework begins with Laplacian mode computation and the approximation of a computationally low-cost Gaussian process surrogate model to capture the minimum weighted combination of inlet-outlet pressure drop (PD) and maximum wall shear stress (WSS). Bayesian Optimization then performs a limited number of OpenFOAM simulations to identify the optimal patient-specific shape.</div><div>We use imaging and flow data obtained from six patients diagnosed with cognitional heart disease to evaluate our approach. Our results showcase the potential of online training and hemodynamic surrogate model optimization for providing optimal graft shapes. These results show how our framework successfully reduces inlet-outlet PD and maximum WSS compared to pre-lofted models that include both the native geometry and human-designed grafts. Furthermore, we compare how the performance of each design optimized under steady-state simulation compares to that design’s performance under transient simulation, and to what extent the optimal design remains similar under both conditions. Our findings underscore that the automated designs achieve at least a 16% reduction in blood flow pressure drop in comparison to geometries optimized by humans.</div></div>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"Article 109308"},"PeriodicalIF":7.0,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142663793","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-18DOI: 10.1016/j.compbiomed.2024.109421
Fakher Frikha, Sami Aifa
N-palmitoylethanolamine (PEA) is an endogenous bioactive compound recognized for its anti-inflammatory effects and its role in tissue protection and repair. Despite the proposal of peroxisome proliferator-activated receptor alpha (PPARα) as a potential receptor for PEA, direct evidence of binding remains insufficient. This study offers a comprehensive analysis of human nuclear receptors (NRs) through structural bioinformatics and molecular docking, evaluating a total of 367 unique NR structures across 47 subfamilies. To explore the stability and binding affinity of PEA with selected nuclear receptors, we conducted molecular dynamics simulations following initial docking assessments. The results revealed Hepatocyte Nuclear Factor 4-alpha (HNF4α) as the highest-ranking receptor with a global score of 0.884, closely followed by Hepatocyte Nuclear Factor 4-gamma (HNF4γ) at 0.871 and Retinoic Acid Receptor gamma-1 (RARγ-1) at 0.829. Among these, HNF4γ demonstrated the strongest affinity for PEA, supported by consistent simulation results. In contrast, the PPARα receptor ranked 44th with a global score of 0.519, indicating that PEA may engage more effectively with other nuclear receptors. In conclusion, this study underscores PEA's potential as a multi-target therapeutic agent through its interactions with various nuclear receptors, particularly HNF4γ and the Mineralocorticoid Receptor (MR). The ability of PEA to influence multiple signaling pathways suggests its promise in addressing complex diseases associated with inflammation and metabolic disorders. Additionally, the integration of Root Mean Square Deviation (RMSD) and Gibbs free energy (ΔG) analyses further elucidates the stability and binding affinities of PEA, providing a foundation for future research into its therapeutic applications.
N-棕榈酰乙醇胺(PEA)是一种内源性生物活性化合物,因其抗炎作用及其在组织保护和修复中的作用而得到公认。尽管有人提出过氧化物酶体增殖激活受体α(PPARα)是 PEA 的潜在受体,但直接的结合证据仍然不足。本研究通过结构生物信息学和分子对接对人类核受体(NRs)进行了全面分析,评估了 47 个亚科共 367 种独特的 NR 结构。为了探索 PEA 与选定核受体的稳定性和结合亲和力,我们在初步对接评估后进行了分子动力学模拟。结果显示,肝细胞核因子 4-α(HNF4α)是全局得分最高的受体,为 0.884,紧随其后的是肝细胞核因子 4-γ(HNF4γ),为 0.871,以及视黄酸受体γ-1(RARγ-1),为 0.829。在这些受体中,HNF4γ 对 PEA 的亲和力最强,这与模拟结果一致。相比之下,PPARα受体以 0.519 的总分排名第 44 位,这表明 PEA 与其他核受体的接触可能更有效。总之,本研究强调了 PEA 通过与各种核受体(尤其是 HNF4γ 和矿质皮质激素受体 (MR))相互作用而成为多靶点治疗药物的潜力。PEA 影响多种信号通路的能力表明,它有望解决与炎症和代谢紊乱有关的复杂疾病。此外,均方根偏差(RMSD)和吉布斯自由能(ΔG)分析的整合进一步阐明了 PEA 的稳定性和结合亲和力,为今后的治疗应用研究奠定了基础。
{"title":"Evaluation of N-palmitoylethanolamine (PEA) binding to nuclear receptors through docking and molecular dynamics studies.","authors":"Fakher Frikha, Sami Aifa","doi":"10.1016/j.compbiomed.2024.109421","DOIUrl":"https://doi.org/10.1016/j.compbiomed.2024.109421","url":null,"abstract":"<p><p>N-palmitoylethanolamine (PEA) is an endogenous bioactive compound recognized for its anti-inflammatory effects and its role in tissue protection and repair. Despite the proposal of peroxisome proliferator-activated receptor alpha (PPARα) as a potential receptor for PEA, direct evidence of binding remains insufficient. This study offers a comprehensive analysis of human nuclear receptors (NRs) through structural bioinformatics and molecular docking, evaluating a total of 367 unique NR structures across 47 subfamilies. To explore the stability and binding affinity of PEA with selected nuclear receptors, we conducted molecular dynamics simulations following initial docking assessments. The results revealed Hepatocyte Nuclear Factor 4-alpha (HNF4α) as the highest-ranking receptor with a global score of 0.884, closely followed by Hepatocyte Nuclear Factor 4-gamma (HNF4γ) at 0.871 and Retinoic Acid Receptor gamma-1 (RARγ-1) at 0.829. Among these, HNF4γ demonstrated the strongest affinity for PEA, supported by consistent simulation results. In contrast, the PPARα receptor ranked 44th with a global score of 0.519, indicating that PEA may engage more effectively with other nuclear receptors. In conclusion, this study underscores PEA's potential as a multi-target therapeutic agent through its interactions with various nuclear receptors, particularly HNF4γ and the Mineralocorticoid Receptor (MR). The ability of PEA to influence multiple signaling pathways suggests its promise in addressing complex diseases associated with inflammation and metabolic disorders. Additionally, the integration of Root Mean Square Deviation (RMSD) and Gibbs free energy (ΔG) analyses further elucidates the stability and binding affinities of PEA, providing a foundation for future research into its therapeutic applications.</p>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"109421"},"PeriodicalIF":7.0,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bacillus anthracis, a gram-positive bacillus capable of forming spores, causes anthrax in mammals, including humans, and is recognized as a potential biological weapon agent. The diagnosis of anthrax is challenging due to variable symptoms resulting from exposure and infection severity. Despite the availability of a licensed vaccines, their limited long-term efficacy underscores the inadequacy of current human anthrax vaccines, highlighting the urgent need for next-generation alternatives. Our study aimed to identify molecular biomarkers and essential biological pathways for the early detection and accurate diagnosis of human anthrax infection. Using a comparative analysis of Bacillus anthracis gene expression data from the Gene Expression Omnibus (GEO) database, this cost-effective approach enables the identification of shared differentially expressed genes (DEGs) across separate microarray datasets without additional hybridization. Three microarray datasets (GSE34407, GSE14390, and GSE12131) of B. anthracis-infected human cell lines were analyzed via the GEO2R tool to identify shared DEGs. We identified 241 common DEGs (70 upregulated and 171 downregulated) from cell lines treated similarly to lethal toxins. Additionally, 10 common DEGs (5 upregulated and 5 downregulated) were identified across different treatments (lethal toxins and spores) and cell lines. Network meta-analysis identified JUN and GATAD2A as the top hub genes for overexpression, and NEDD4L and GULP1 for underexpression. Furthermore, prognostic analysis and SNP detection of the two identified upregulated hub genes were carried out in conjunction with machine learning classification models, with SVM yielding the best classification accuracy of 87.5 %. Our comparative analysis of Bacillus anthracis infection revealed striking similarities in gene expression 241 profiles across diverse datasets, despite variations in treatments and cell lines. These findings underscore how anthrax infection activates shared genes across different cell types, emphasizing this approach in the discovery of novel gene markers. These markers offer insights into pathogenesis and may lead to more effective therapeutic strategies. By identifying these genetic indicators, we can advance the development of precise immunotherapies, potentially enhancing vaccine efficacy and treatment outcomes.
{"title":"Identification of molecular and cellular infection response biomarkers associated with anthrax infection through comparative analysis of gene expression data","authors":"Swati Rani , Varsha Ramesh , Mehnaj Khatoon , M. Shijili , C.A. Archana , Jayashree Anand , N. Sagar , Yamini S. Sekar , Archana V. Patil , Azhahianambi Palavesam , N.N. Barman , S.S. Patil , Diwakar Hemadri , K.P. Suresh","doi":"10.1016/j.compbiomed.2024.109431","DOIUrl":"10.1016/j.compbiomed.2024.109431","url":null,"abstract":"<div><div><em>Bacillus anthracis,</em> a gram-positive bacillus capable of forming spores, causes anthrax in mammals, including humans, and is recognized as a potential biological weapon agent. The diagnosis of anthrax is challenging due to variable symptoms resulting from exposure and infection severity. Despite the availability of a licensed vaccines, their limited long-term efficacy underscores the inadequacy of current human anthrax vaccines, highlighting the urgent need for next-generation alternatives. Our study aimed to identify molecular biomarkers and essential biological pathways for the early detection and accurate diagnosis of human anthrax infection. Using a comparative analysis of <em>Bacillus anthracis</em> gene expression data from the Gene Expression Omnibus (GEO) database, this cost-effective approach enables the identification of shared differentially expressed genes (DEGs) across separate microarray datasets without additional hybridization. Three microarray datasets (GSE34407, GSE14390, and GSE12131) of <em>B. anthracis</em>-infected human cell lines were analyzed via the GEO2R tool to identify shared DEGs. We identified 241 common DEGs (70 upregulated and 171 downregulated) from cell lines treated similarly to lethal toxins. Additionally, 10 common DEGs (5 upregulated and 5 downregulated) were identified across different treatments (lethal toxins and spores) and cell lines. Network meta-analysis identified <em>JUN</em> and <em>GATAD2A</em> as the top hub genes for overexpression, and <em>NEDD4L</em> and <em>GULP1</em> for underexpression. Furthermore, prognostic analysis and SNP detection of the two identified upregulated hub genes were carried out in conjunction with machine learning classification models, with SVM yielding the best classification accuracy of 87.5 %. Our comparative analysis of <em>Bacillus anthracis</em> infection revealed striking similarities in gene expression 241 profiles across diverse datasets, despite variations in treatments and cell lines. These findings underscore how anthrax infection activates shared genes across different cell types, emphasizing this approach in the discovery of novel gene markers. These markers offer insights into pathogenesis and may lead to more effective therapeutic strategies. By identifying these genetic indicators, we can advance the development of precise immunotherapies, potentially enhancing vaccine efficacy and treatment outcomes.</div></div>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"Article 109431"},"PeriodicalIF":7.0,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142663791","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-17DOI: 10.1016/j.compbiomed.2024.109411
SungHwan Moon, Junhyeok Lee, Won Hee Lee
Brain age, an emerging biomarker for brain diseases and aging, is typically predicted using single-modality T1-weighted structural MRI data. This study investigates the benefits of integrating structural MRI with diffusion MRI to enhance brain age prediction. We propose an attention-based deep learning model that fuses global-context information from structural MRI with local details from diffusion metrics. The model was evaluated using two large datasets: the Human Connectome Project (HCP, n = 1064, age 22–37) and the Cambridge Center for Aging and Neuroscience (Cam-CAN, n = 639, age 18–88). It was tested for generalizability and robustness on three independent datasets (n = 546, age 20–86), reproducibility on a test-retest dataset (n = 44, age 22–35), and longitudinal consistency (n = 129, age 46–92). We also examined the relationship between predicted brain age and behavioral measures. Results showed that the multimodal model improved prediction accuracy, achieving mean absolute errors (MAEs) of 2.44 years in the HCP dataset (sagittal plane) and 4.36 years in the Cam-CAN dataset (axial plane). The corresponding R2 values were 0.258 and 0.914, respectively, reflecting the model's ability to explain variance in the predictions across both datasets. Compared to single-modality models, the multimodal approach showed better generalization, reducing MAEs by 10–76 % and enhancing robustness by 22–82 %. While the multimodal model exhibited superior reproducibility, the sMRI model showed slightly better longitudinal consistency. Importantly, the multimodal model revealed unique associations between predicted brain age and behavioral measures, such as walking endurance and loneliness in the HCP dataset, which were not detected with chronological age alone. In the Cam-CAN dataset, brain age and chronological age exhibited similar correlations with behavioral measures. By integrating sMRI and dMRI through an attention-based model, our proposed approach enhances predictive accuracy and provides deeper insights into the relationship between brain aging and behavior.
{"title":"Predicting brain age with global-local attention network from multimodal neuroimaging data: Accuracy, generalizability, and behavioral associations","authors":"SungHwan Moon, Junhyeok Lee, Won Hee Lee","doi":"10.1016/j.compbiomed.2024.109411","DOIUrl":"10.1016/j.compbiomed.2024.109411","url":null,"abstract":"<div><div>Brain age, an emerging biomarker for brain diseases and aging, is typically predicted using single-modality T1-weighted structural MRI data. This study investigates the benefits of integrating structural MRI with diffusion MRI to enhance brain age prediction. We propose an attention-based deep learning model that fuses global-context information from structural MRI with local details from diffusion metrics. The model was evaluated using two large datasets: the Human Connectome Project (HCP, n = 1064, age 22–37) and the Cambridge Center for Aging and Neuroscience (Cam-CAN, n = 639, age 18–88). It was tested for generalizability and robustness on three independent datasets (n = 546, age 20–86), reproducibility on a test-retest dataset (n = 44, age 22–35), and longitudinal consistency (n = 129, age 46–92). We also examined the relationship between predicted brain age and behavioral measures. Results showed that the multimodal model improved prediction accuracy, achieving mean absolute errors (MAEs) of 2.44 years in the HCP dataset (sagittal plane) and 4.36 years in the Cam-CAN dataset (axial plane). The corresponding R<sup>2</sup> values were 0.258 and 0.914, respectively, reflecting the model's ability to explain variance in the predictions across both datasets. Compared to single-modality models, the multimodal approach showed better generalization, reducing MAEs by 10–76 % and enhancing robustness by 22–82 %. While the multimodal model exhibited superior reproducibility, the sMRI model showed slightly better longitudinal consistency. Importantly, the multimodal model revealed unique associations between predicted brain age and behavioral measures, such as walking endurance and loneliness in the HCP dataset, which were not detected with chronological age alone. In the Cam-CAN dataset, brain age and chronological age exhibited similar correlations with behavioral measures. By integrating sMRI and dMRI through an attention-based model, our proposed approach enhances predictive accuracy and provides deeper insights into the relationship between brain aging and behavior.</div></div>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"Article 109411"},"PeriodicalIF":7.0,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142663790","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-17DOI: 10.1016/j.compbiomed.2024.109419
Tatsuya Tanaka , Toshiaki Katayama , Takeshi Imai
Background
Multiple studies have aimed to consolidate drug-related data and predict drug effects. However, most of these studies have focused on integrating diverse data through correlation rather than representing them based on the pharmacodynamic mechanism of action (MOA). It is thus crucial to obtain interpretability to validate prediction results. In this study, we propose a novel framework to construct knowledge graphs that represent pharmacodynamic MOA, predict drug effects, and derive conceivable mechanistic pathways.
Methods and results
We constructed an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG) by integrating various existing databases and combining them with the approach of this study to automatically fill in the missing data. This yielded a knowledge graph comprising 1455 drugs and 2547 diseases. Additionally, a graph neural network (GNN)-based approach was used to predict therapeutic medication and indication, which outperformed previous approaches that relied on correlation-based knowledge graphs lacking pharmacodynamic MOA representations. Furthermore, we proposed and assessed a method to interpret pharmacodynamic MOA using gene perturbation data. This feasibility study demonstrated the successful derivation of an accurate mechanism in approximately 50 % of cases. Additionally, it facilitated the identification of candidate drugs, which are currently unapproved but exhibit potential for drug repositioning, and their mechanisms of action.
Conclusions
This framework not only enables the derivation of highly accurate “drug–indication” predictions but also offers a basic mechanistic understanding, thereby facilitating future drug repositioning efforts.
{"title":"Predicting the effects of drugs and unveiling their mechanisms of action using an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG)","authors":"Tatsuya Tanaka , Toshiaki Katayama , Takeshi Imai","doi":"10.1016/j.compbiomed.2024.109419","DOIUrl":"10.1016/j.compbiomed.2024.109419","url":null,"abstract":"<div><h3>Background</h3><div>Multiple studies have aimed to consolidate drug-related data and predict drug effects. However, most of these studies have focused on integrating diverse data through correlation rather than representing them based on the pharmacodynamic mechanism of action (MOA). It is thus crucial to obtain interpretability to validate prediction results. In this study, we propose a novel framework to construct knowledge graphs that represent pharmacodynamic MOA, predict drug effects, and derive conceivable mechanistic pathways.</div></div><div><h3>Methods and results</h3><div>We constructed an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG) by integrating various existing databases and combining them with the approach of this study to automatically fill in the missing data. This yielded a knowledge graph comprising 1455 drugs and 2547 diseases. Additionally, a graph neural network (GNN)-based approach was used to predict therapeutic medication and indication, which outperformed previous approaches that relied on correlation-based knowledge graphs lacking pharmacodynamic MOA representations. Furthermore, we proposed and assessed a method to interpret pharmacodynamic MOA using gene perturbation data. This feasibility study demonstrated the successful derivation of an accurate mechanism in approximately 50 % of cases. Additionally, it facilitated the identification of candidate drugs, which are currently unapproved but exhibit potential for drug repositioning, and their mechanisms of action.</div></div><div><h3>Conclusions</h3><div>This framework not only enables the derivation of highly accurate “drug–indication” predictions but also offers a basic mechanistic understanding, thereby facilitating future drug repositioning efforts.</div></div>","PeriodicalId":10578,"journal":{"name":"Computers in biology and medicine","volume":"184 ","pages":"Article 109419"},"PeriodicalIF":7.0,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142663792","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}