Pub Date : 2024-11-09DOI: 10.1038/s41540-024-00464-z
Xuefei Lin, Xiao Chang, Yizheng Zhang, Zhanyu Gao, Xu Chi
Petri nets are commonly applied in modeling biological systems. However, construction of a Petri net model for complex biological systems is often time consuming, and requires expertise in the research area, limiting their application. To address this challenge, we developed GINtoSPN, an R package that automates the conversion of multi-omics molecular interaction network extracted from the Global Integrative Network (GIN) into Petri nets in GraphML format. These GraphML files can be directly used for Signaling Petri Net (SPN) simulation. To demonstrate the utility of this tool, we built a Petri net model for neurofibromatosis type I. Simulation of NF1 gene knockout, compared to normal skin fibroblast cells, revealed persistent accumulation of Ras-GTPs as expected. Additionally, we identified several other genes substantially affected by the loss of NF1's function, exhibiting individual-specific variability. These results highlight the effectiveness of GINtoSPN in streamlining the modeling and simulation of complex biological systems.
Petri 网通常用于生物系统建模。然而,为复杂的生物系统构建 Petri 网模型往往非常耗时,而且需要研究领域的专业知识,这限制了 Petri 网的应用。为了应对这一挑战,我们开发了 GINtoSPN 这个 R 软件包,它能自动将从全球整合网络(GIN)中提取的多组学分子相互作用网络转换成 GraphML 格式的 Petri 网。这些 GraphML 文件可直接用于信号 Petri 网(SPN)模拟。为了证明这一工具的实用性,我们为 I 型神经纤维瘤病建立了一个 Petri 网模型。与正常皮肤成纤维细胞相比,NF1 基因敲除的模拟结果显示,Ras-GTPs 的持续积累符合预期。此外,我们还发现了其他几个基因因 NF1 功能缺失而受到严重影响,并表现出个体特异性。这些结果凸显了 GINtoSPN 在简化复杂生物系统建模和仿真方面的有效性。
{"title":"Automatic construction of Petri net models for computational simulations of molecular interaction network.","authors":"Xuefei Lin, Xiao Chang, Yizheng Zhang, Zhanyu Gao, Xu Chi","doi":"10.1038/s41540-024-00464-z","DOIUrl":"10.1038/s41540-024-00464-z","url":null,"abstract":"<p><p>Petri nets are commonly applied in modeling biological systems. However, construction of a Petri net model for complex biological systems is often time consuming, and requires expertise in the research area, limiting their application. To address this challenge, we developed GINtoSPN, an R package that automates the conversion of multi-omics molecular interaction network extracted from the Global Integrative Network (GIN) into Petri nets in GraphML format. These GraphML files can be directly used for Signaling Petri Net (SPN) simulation. To demonstrate the utility of this tool, we built a Petri net model for neurofibromatosis type I. Simulation of NF1 gene knockout, compared to normal skin fibroblast cells, revealed persistent accumulation of Ras-GTPs as expected. Additionally, we identified several other genes substantially affected by the loss of NF1's function, exhibiting individual-specific variability. These results highlight the effectiveness of GINtoSPN in streamlining the modeling and simulation of complex biological systems.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"131"},"PeriodicalIF":3.5,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550427/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142624958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Since nail psoriasis restricts the patient's daily activities, therapeutic intervention based on reliable and reproducible evaluation is critical. The Nail Psoriasis Severity Index (NAPSI) is a validated scoring tool, but its usefulness is limited by interobserver variability. This study aimed to develop a reliable and accurate NAPSI scoring tool using deep learning. The tool "NAPSI calculator" includes two parts: nail detection from images and NAPSI scoring. NAPSI was annotated by nine nail experts who are board-certified dermatologists with sufficient experience in a specialized clinic for nail diseases. In the final test set, the "NAPSI calculator" correctly located 137/138 nails and scored NAPSI with higher accuracy than the compared six non-board-certified residents: 83.9% vs 65.7%; P = 0.008 and four board-certified non-nail expert dermatologists: 83.9% vs 73.0%; P = 0.005. The "NAPSI calculator" can be readily used in a clinical situation, contributing to raising the medical practice level for nail psoriasis.
由于指甲银屑病会限制患者的日常活动,因此基于可靠、可重复评估的治疗干预至关重要。指甲银屑病严重程度指数(NAPSI)是一种经过验证的评分工具,但其实用性受到观察者之间差异性的限制。本研究旨在利用深度学习技术开发一种可靠、准确的 NAPSI 评分工具。该工具 "NAPSI计算器 "包括两部分:从图像中检测指甲和NAPSI评分。NAPSI 由九名美甲专家进行注释,他们都是在甲病专科门诊工作过并拥有丰富经验的注册皮肤科医生。在最终测试集中,"NAPSI 计算器 "正确定位了 137/138 枚指甲,并对 NAPSI 进行了评分,准确率高于六位未获得医学会认证的住院医师:83.9% vs 65.7%; P = 0.008,以及四位获得医学会认证的非指甲专家皮肤科医生:83.9% vs 73.0%; P = 0.005。NAPSI 计算器 "可随时用于临床,有助于提高甲银屑病的医疗实践水平。
{"title":"Reliable and easy-to-use calculating tool for the Nail Psoriasis Severity Index using deep learning.","authors":"Hiroto Horikawa, Keiji Tanese, Naoki Nonaka, Jun Seita, Masayuki Amagai, Masataka Saito","doi":"10.1038/s41540-024-00458-x","DOIUrl":"10.1038/s41540-024-00458-x","url":null,"abstract":"<p><p>Since nail psoriasis restricts the patient's daily activities, therapeutic intervention based on reliable and reproducible evaluation is critical. The Nail Psoriasis Severity Index (NAPSI) is a validated scoring tool, but its usefulness is limited by interobserver variability. This study aimed to develop a reliable and accurate NAPSI scoring tool using deep learning. The tool \"NAPSI calculator\" includes two parts: nail detection from images and NAPSI scoring. NAPSI was annotated by nine nail experts who are board-certified dermatologists with sufficient experience in a specialized clinic for nail diseases. In the final test set, the \"NAPSI calculator\" correctly located 137/138 nails and scored NAPSI with higher accuracy than the compared six non-board-certified residents: 83.9% vs 65.7%; P = 0.008 and four board-certified non-nail expert dermatologists: 83.9% vs 73.0%; P = 0.005. The \"NAPSI calculator\" can be readily used in a clinical situation, contributing to raising the medical practice level for nail psoriasis.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"130"},"PeriodicalIF":4.3,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544089/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142605120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Understanding the dynamic states and transitions of heterogeneous cell populations is crucial for addressing fundamental biological questions. High-content imaging provides rich datasets, but it remains increasingly difficult to integrate and annotate high-dimensional and time-resolved datasets to profile heterogeneous cell population dynamics in different microenvironments. Using hepatic stellate cells (HSCs) LX-2 as model, we proposed a novel analytical strategy for image-based integration and annotation to profile dynamics of heterogeneous cell populations in 2D/3D microenvironments. High-dimensional features were extracted from extensive image datasets, and cellular states were identified based on feature profiles. Time-series clustering revealed distinct temporal patterns of cell shape and actin cytoskeleton reorganization. We found LX-2 showed more complex membrane dynamics and contractile systems with an M-shaped actin compactness trend in 3D culture, while they displayed rapid spreading in early 2D culture. This image-based integration and annotation strategy enhances our understanding of HSCs heterogeneity and dynamics in complex extracellular microenvironments.
了解异质细胞群的动态状态和转变对于解决基本生物学问题至关重要。高含量成像技术提供了丰富的数据集,但要整合和注释高维和时间分辨数据集以剖析不同微环境中异质细胞群的动态变化却越来越难。我们以肝星状细胞(HSCs)LX-2为模型,提出了一种基于图像整合和注释的新型分析策略,以剖析2D/3D微环境中异质细胞群的动态。我们从大量图像数据集中提取了高维特征,并根据特征轮廓确定了细胞状态。时间序列聚类揭示了细胞形状和肌动蛋白细胞骨架重组的独特时间模式。我们发现,LX-2 在三维培养中表现出更复杂的膜动力学和收缩系统,其肌动蛋白紧密度呈 M 型趋势,而在早期二维培养中则表现出快速扩散。这种基于图像的整合和注释策略增强了我们对造血干细胞在复杂细胞外微环境中异质性和动态性的理解。
{"title":"Exploring heterogeneous cell population dynamics in different microenvironments by novel analytical strategy based on images.","authors":"Yihong Huang, Zidong Zhou, Tianqi Liu, Shengnan Tang, Xuegang Xin","doi":"10.1038/s41540-024-00459-w","DOIUrl":"10.1038/s41540-024-00459-w","url":null,"abstract":"<p><p>Understanding the dynamic states and transitions of heterogeneous cell populations is crucial for addressing fundamental biological questions. High-content imaging provides rich datasets, but it remains increasingly difficult to integrate and annotate high-dimensional and time-resolved datasets to profile heterogeneous cell population dynamics in different microenvironments. Using hepatic stellate cells (HSCs) LX-2 as model, we proposed a novel analytical strategy for image-based integration and annotation to profile dynamics of heterogeneous cell populations in 2D/3D microenvironments. High-dimensional features were extracted from extensive image datasets, and cellular states were identified based on feature profiles. Time-series clustering revealed distinct temporal patterns of cell shape and actin cytoskeleton reorganization. We found LX-2 showed more complex membrane dynamics and contractile systems with an M-shaped actin compactness trend in 3D culture, while they displayed rapid spreading in early 2D culture. This image-based integration and annotation strategy enhances our understanding of HSCs heterogeneity and dynamics in complex extracellular microenvironments.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"129"},"PeriodicalIF":3.5,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11542073/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142591310","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-05DOI: 10.1038/s41540-024-00449-y
Mucen Yu, Jielin Xu, Ranjan Dutta, Bruce Trapp, Andrew A Pieper, Feixiong Cheng
Amyotrophic Lateral Sclerosis (ALS) is a devastating, immensely complex neurodegenerative disease by lack of effective treatments. We developed a network medicine methodology via integrating human brain multi-omics data to prioritize drug targets and repurposable treatments for ALS. We leveraged non-coding ALS loci effects from genome-wide associated studies (GWAS) on human brain expression quantitative trait loci (QTL) (eQTL), protein QTL (pQTL), splicing QTL (sQTL), methylation QTL (meQTL), and histone acetylation QTL (haQTL). Using a network-based deep learning framework, we identified 105 putative ALS-associated genes enriched in known ALS pathobiological pathways. Applying network proximity analysis of predicted ALS-associated genes and drug-target networks under the human protein-protein interactome (PPI) model, we identified potential repurposable drugs (i.e., Diazoxide and Gefitinib) for ALS. Subsequent validation established preclinical evidence for top-prioritized drugs. In summary, we presented a network-based multi-omics framework to identify drug targets and repurposable treatments for ALS and other neurodegenerative disease if broadly applied.
肌萎缩性脊髓侧索硬化症(ALS)是一种毁灭性的、极其复杂的神经退行性疾病,缺乏有效的治疗方法。我们开发了一种网络医学方法,通过整合人脑多组学数据来优先确定 ALS 的药物靶点和可再利用的治疗方法。我们利用全基因组关联研究(GWAS)中关于人脑表达定量性状位点(QTL)(eQTL)、蛋白质定量性状位点(pQTL)、剪接定量性状位点(sQTL)、甲基化定量性状位点(meQTL)和组蛋白乙酰化定量性状位点(haQTL)的非编码 ALS 位点效应。利用基于网络的深度学习框架,我们在已知的 ALS 病理生物学通路中发现了 105 个假定的 ALS 相关基因。通过对预测的 ALS 相关基因和人类蛋白质-蛋白质相互作用组(PPI)模型下的药物-靶点网络进行网络邻近性分析,我们确定了治疗 ALS 的潜在可再利用药物(即 Diazoxide 和 Gefitinib)。随后的验证为优先药物提供了临床前证据。总之,我们提出了一个基于网络的多组学框架,以确定药物靶点,并在广泛应用的情况下确定可再利用的治疗 ALS 和其他神经退行性疾病的方法。
{"title":"Network medicine informed multiomics integration identifies drug targets and repurposable medicines for Amyotrophic Lateral Sclerosis.","authors":"Mucen Yu, Jielin Xu, Ranjan Dutta, Bruce Trapp, Andrew A Pieper, Feixiong Cheng","doi":"10.1038/s41540-024-00449-y","DOIUrl":"10.1038/s41540-024-00449-y","url":null,"abstract":"<p><p>Amyotrophic Lateral Sclerosis (ALS) is a devastating, immensely complex neurodegenerative disease by lack of effective treatments. We developed a network medicine methodology via integrating human brain multi-omics data to prioritize drug targets and repurposable treatments for ALS. We leveraged non-coding ALS loci effects from genome-wide associated studies (GWAS) on human brain expression quantitative trait loci (QTL) (eQTL), protein QTL (pQTL), splicing QTL (sQTL), methylation QTL (meQTL), and histone acetylation QTL (haQTL). Using a network-based deep learning framework, we identified 105 putative ALS-associated genes enriched in known ALS pathobiological pathways. Applying network proximity analysis of predicted ALS-associated genes and drug-target networks under the human protein-protein interactome (PPI) model, we identified potential repurposable drugs (i.e., Diazoxide and Gefitinib) for ALS. Subsequent validation established preclinical evidence for top-prioritized drugs. In summary, we presented a network-based multi-omics framework to identify drug targets and repurposable treatments for ALS and other neurodegenerative disease if broadly applied.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"128"},"PeriodicalIF":3.5,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11538253/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142583433","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04DOI: 10.1038/s41540-024-00455-0
Gøran Troseth Andersen, Aleksandr Ianevski, Mathilde Resell, Naris Pojskic, Hanne-Line Rabben, Synne Geithus, Yosuke Kodama, Tomita Hiroyuki, Denis Kainov, Jon Erik Grønbech, Yoku Hayakawa, Timothy C Wang, Chun-Mei Zhao, Duan Chen
Biomarkers associated with the progression from gastric intestinal metaplasia (GIM) to gastric adenocarcinoma (GA), i.e., GA-related GIM, could provide valuable insights into identifying patients with increased risk for GA. The aim of this study was to utilize multi-bioinformatics to reveal potential biomarkers for the GA-related GIM and predict potential drug repurposing for GA prevention in patients. The multi-bioinformatics included gene expression matrix (GEM) by microarray gene expression (MGE), ScType (a fully automated and ultra-fast cell-type identification based solely on a given scRNA-seq data), Ingenuity Pathway Analysis, PageRank centrality, GO and MSigDB enrichments, Cytoscape, Human Protein Atlas and molecular docking analysis in combination with immunohistochemistry. To identify GA-related GIM, paired surgical biopsies were collected from 16 GIM-GA patients who underwent gastrectomy, yielding 64 samples (4 biopsies per stomach x 16 patients) for MGE. Co-analysis was performed by including scRNAseq and immunohistochemistry datasets of endoscopic biopsies of 37 patients. The results of the present study showed potential biomarkers for GA-related GIM, including GEM of individual patients, individual genes (such as RBP2 and CD44), signaling pathways, network of molecules, and network of signaling pathways with key topological nodes. Accordingly, potential treatment targets with repurposed drugs were identified including epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase Src, paxillin, transcription factor Jun, breast cancer type 1 susceptibility protein, cellular tumor antigen p53, mouse double minute 2, and CD44.
与胃肠化生(GIM)发展为胃腺癌(GA)相关的生物标志物,即与GA相关的GIM,可为识别GA风险增加的患者提供有价值的见解。本研究旨在利用多元生物信息学揭示与 GA 相关的 GIM 的潜在生物标志物,并预测预防患者 GA 的潜在药物再利用。多元生物信息学包括微阵列基因表达(MGE)的基因表达矩阵(GEM)、ScType(一种完全基于给定scRNA-seq数据的全自动超快速细胞类型鉴定)、Ingenuity Pathway Analysis、PageRank centrality、GO和MSigDB富集、Cytoscape、Human Protein Atlas以及结合免疫组化的分子对接分析。为确定与 GA 相关的 GIM,从 16 名接受胃切除术的 GIM-GA 患者身上收集了配对的手术活检样本,共获得 64 个 MGE 样本(每个胃 4 个活检样本 x 16 名患者)。通过纳入 37 例患者内镜活检的 scRNAseq 和免疫组化数据集进行了联合分析。本研究的结果显示了GA相关GIM的潜在生物标记物,包括单个患者的GEM、单个基因(如RBP2和CD44)、信号通路、分子网络和具有关键拓扑节点的信号通路网络。据此,研究发现了表皮生长因子受体、原癌基因酪氨酸蛋白激酶Src、paxillin、转录因子Jun、乳腺癌1型易感蛋白、细胞肿瘤抗原p53、小鼠双分化2和CD44等再利用药物的潜在治疗靶点。
{"title":"Multi-bioinformatics revealed potential biomarkers and repurposed drugs for gastric adenocarcinoma-related gastric intestinal metaplasia.","authors":"Gøran Troseth Andersen, Aleksandr Ianevski, Mathilde Resell, Naris Pojskic, Hanne-Line Rabben, Synne Geithus, Yosuke Kodama, Tomita Hiroyuki, Denis Kainov, Jon Erik Grønbech, Yoku Hayakawa, Timothy C Wang, Chun-Mei Zhao, Duan Chen","doi":"10.1038/s41540-024-00455-0","DOIUrl":"10.1038/s41540-024-00455-0","url":null,"abstract":"<p><p>Biomarkers associated with the progression from gastric intestinal metaplasia (GIM) to gastric adenocarcinoma (GA), i.e., GA-related GIM, could provide valuable insights into identifying patients with increased risk for GA. The aim of this study was to utilize multi-bioinformatics to reveal potential biomarkers for the GA-related GIM and predict potential drug repurposing for GA prevention in patients. The multi-bioinformatics included gene expression matrix (GEM) by microarray gene expression (MGE), ScType (a fully automated and ultra-fast cell-type identification based solely on a given scRNA-seq data), Ingenuity Pathway Analysis, PageRank centrality, GO and MSigDB enrichments, Cytoscape, Human Protein Atlas and molecular docking analysis in combination with immunohistochemistry. To identify GA-related GIM, paired surgical biopsies were collected from 16 GIM-GA patients who underwent gastrectomy, yielding 64 samples (4 biopsies per stomach x 16 patients) for MGE. Co-analysis was performed by including scRNAseq and immunohistochemistry datasets of endoscopic biopsies of 37 patients. The results of the present study showed potential biomarkers for GA-related GIM, including GEM of individual patients, individual genes (such as RBP2 and CD44), signaling pathways, network of molecules, and network of signaling pathways with key topological nodes. Accordingly, potential treatment targets with repurposed drugs were identified including epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase Src, paxillin, transcription factor Jun, breast cancer type 1 susceptibility protein, cellular tumor antigen p53, mouse double minute 2, and CD44.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"127"},"PeriodicalIF":3.5,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11535201/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142576710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disease that affects about 0.1% to 2% of the population worldwide. Despite the development of several novel therapies, there is only limited benefit for many patients. Thus, there is room for new approaches to improve response to therapy, including designing better trials e.g., by identifying subpopulations that can benefit from specific classes of therapy and enabling reverse translation by analyzing completed clinical trials. We have developed an open-source, mechanistic multi-scale model of RA, which captures the interactions of key immune cells and mediators in an inflamed joint. The model consists of a treatment-naive Virtual Population (Vpop) that responds appropriately (i.e. as reported in clinical trials) to standard-of-care treatment options-Methotrexate (MTX) and Adalimumab (ADA, anti-TNF-α) and an MTX inadequate responder sub-population that responds appropriately to Tocilizumab (TCZ, anti-IL-6R) therapy. The clinical read-outs of interest are the American College of Rheumatology score (ACR score) and Disease Activity Score (DAS28-CRP), which is modeled to be dependent on the physiological variables in the model. Further, we have validated the Vpop by predicting the therapy response of TCZ on ADA Non-responders. This paper aims to share our approach, equations, and code to enable community evaluation and greater adoption of mechanistic models in drug development for autoimmune diseases.
{"title":"Multiscale, mechanistic model of Rheumatoid Arthritis to enable decision making in late stage drug development.","authors":"Dinesh Bedathuru, Maithreye Rengaswamy, Madhav Channavazzala, Tamara Ray, Prakash Packrisamy, Rukmini Kumar","doi":"10.1038/s41540-024-00454-1","DOIUrl":"10.1038/s41540-024-00454-1","url":null,"abstract":"<p><p>Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disease that affects about 0.1% to 2% of the population worldwide. Despite the development of several novel therapies, there is only limited benefit for many patients. Thus, there is room for new approaches to improve response to therapy, including designing better trials e.g., by identifying subpopulations that can benefit from specific classes of therapy and enabling reverse translation by analyzing completed clinical trials. We have developed an open-source, mechanistic multi-scale model of RA, which captures the interactions of key immune cells and mediators in an inflamed joint. The model consists of a treatment-naive Virtual Population (Vpop) that responds appropriately (i.e. as reported in clinical trials) to standard-of-care treatment options-Methotrexate (MTX) and Adalimumab (ADA, anti-TNF-α) and an MTX inadequate responder sub-population that responds appropriately to Tocilizumab (TCZ, anti-IL-6R) therapy. The clinical read-outs of interest are the American College of Rheumatology score (ACR score) and Disease Activity Score (DAS28-CRP), which is modeled to be dependent on the physiological variables in the model. Further, we have validated the Vpop by predicting the therapy response of TCZ on ADA Non-responders. This paper aims to share our approach, equations, and code to enable community evaluation and greater adoption of mechanistic models in drug development for autoimmune diseases.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"126"},"PeriodicalIF":3.5,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11535547/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142576711","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-26DOI: 10.1038/s41540-024-00425-6
Roberta Marino, Yousef El Aalamat, Vanesa Bol, Michele Caselle, Giuseppe Del Giudice, Christophe Lambert, Duccio Medini, Tom M A Wilkinson, Alessandro Muzzi
Chronic obstructive pulmonary disease (COPD) is an etiologically complex disease characterized by acute exacerbations and stable phases. We aimed to identify biological functions modulated in specific COPD conditions, using whole blood samples collected in the AERIS clinical study (NCT01360398). Considered conditions were exacerbation onset, severity of airway obstruction, and presence of respiratory pathogens in sputum samples. With an integrative multi-network gene community detection (MNGCD) approach, we analyzed expression profiles to identify communities of correlated genes. The approach combined different layers of gene interactions for each explored condition/subset of samples: gene expression similarity, protein-protein interactions, transcription factors, and microRNAs validated regulons. Heme metabolism, interferon-alpha, and interferon-gamma pathways were modulated in patients at both exacerbation and stable-state visits, but with the involvement of distinct sets of genes. An important gene community was enriched with G2M checkpoint, E2F targets, and mitotic spindle pathways during exacerbation. Targets of TAL1 regulator and hsa-let-7b - 5p microRNA were modulated with increasing severity of airway obstruction. Bacterial infections with Moraxella catarrhalis and, particularly, Haemophilus influenzae triggered a specific cellular and inflammatory response in acute exacerbations, indicating an active reaction of the host to infections. In conclusion, COPD is a complex multifactorial disease that requires in-depth investigations of its causes and features during its evolution and whole blood transcriptome profiling can contribute to capturing some relevant regulatory mechanisms associated with this disease. In this work, we explored multi-network modeling that integrated diverse layers of regulatory gene networks and enhanced our comprehension of the biological functions implicated in the COPD pathogenesis.
{"title":"An integrative network-based approach to identify driving gene communities in chronic obstructive pulmonary disease.","authors":"Roberta Marino, Yousef El Aalamat, Vanesa Bol, Michele Caselle, Giuseppe Del Giudice, Christophe Lambert, Duccio Medini, Tom M A Wilkinson, Alessandro Muzzi","doi":"10.1038/s41540-024-00425-6","DOIUrl":"10.1038/s41540-024-00425-6","url":null,"abstract":"<p><p>Chronic obstructive pulmonary disease (COPD) is an etiologically complex disease characterized by acute exacerbations and stable phases. We aimed to identify biological functions modulated in specific COPD conditions, using whole blood samples collected in the AERIS clinical study (NCT01360398). Considered conditions were exacerbation onset, severity of airway obstruction, and presence of respiratory pathogens in sputum samples. With an integrative multi-network gene community detection (MNGCD) approach, we analyzed expression profiles to identify communities of correlated genes. The approach combined different layers of gene interactions for each explored condition/subset of samples: gene expression similarity, protein-protein interactions, transcription factors, and microRNAs validated regulons. Heme metabolism, interferon-alpha, and interferon-gamma pathways were modulated in patients at both exacerbation and stable-state visits, but with the involvement of distinct sets of genes. An important gene community was enriched with G2M checkpoint, E2F targets, and mitotic spindle pathways during exacerbation. Targets of TAL1 regulator and hsa-let-7b - 5p microRNA were modulated with increasing severity of airway obstruction. Bacterial infections with Moraxella catarrhalis and, particularly, Haemophilus influenzae triggered a specific cellular and inflammatory response in acute exacerbations, indicating an active reaction of the host to infections. In conclusion, COPD is a complex multifactorial disease that requires in-depth investigations of its causes and features during its evolution and whole blood transcriptome profiling can contribute to capturing some relevant regulatory mechanisms associated with this disease. In this work, we explored multi-network modeling that integrated diverse layers of regulatory gene networks and enhanced our comprehension of the biological functions implicated in the COPD pathogenesis.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"125"},"PeriodicalIF":3.5,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11513021/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Elucidating the emergent dynamics of cellular differentiation networks is crucial to understanding cell-fate decisions. Toggle switch - a network of mutually repressive lineage-specific transcription factors A and B - enables two phenotypes from a common progenitor: (high A, low B) and (low A, high B). However, the dynamics of networks enabling differentiation of more than two phenotypes from a progenitor cell has not been well-studied. Here, we investigate the dynamics of a four-node network A, B, C, and D inhibiting each other, forming a toggle tetrahedron. Our simulations show that this network is multistable and predominantly allows for the co-existence of six hybrid phenotypes where two of the nodes are expressed relatively high as compared to the remaining two, for instance (high A, high B, low C, low D). Finally, we apply our results to understand naïve CD4+ T cell differentiation into Th1, Th2, Th17 and Treg subsets, suggesting Th1/Th2/Th17/Treg decision-making to be a two-step process.
阐明细胞分化网络的新兴动态对于理解细胞命运的决定至关重要。切换开关(Toggle switch)--一个由相互抑制的特异性转录因子 A 和 B 组成的网络--能从一个共同的祖细胞分化出两种表型:(高 A、低 B)和(低 A、高 B)。然而,对能从一个祖细胞分化出两种以上表型的网络的动态还没有进行深入研究。在这里,我们研究了一个四节点网络 A、B、C 和 D 相互抑制,形成一个切换四面体的动力学。我们的模拟结果表明,该网络是多稳态的,主要允许六种混合表型共存,其中两个节点的表达量相对于其余两个节点较高,例如(高 A、高 B、低 C、低 D)。最后,我们将研究结果应用于理解幼稚 CD4+ T 细胞分化成 Th1、Th2、Th17 和 Treg 亚群的过程,这表明 Th1/Th2/Th17/Treg 的决策是一个两步过程。
{"title":"Multistability and predominant hybrid phenotypes in a four node mutually repressive network of Th1/Th2/Th17/Treg differentiation.","authors":"Atchuta Srinivas Duddu, Elizabeth Andreas, Harshavardhan Bv, Kaushal Grover, Vivek Raj Singh, Kishore Hari, Siddharth Jhunjhunwala, Breschine Cummins, Tomas Gedeon, Mohit Kumar Jolly","doi":"10.1038/s41540-024-00433-6","DOIUrl":"https://doi.org/10.1038/s41540-024-00433-6","url":null,"abstract":"<p><p>Elucidating the emergent dynamics of cellular differentiation networks is crucial to understanding cell-fate decisions. Toggle switch - a network of mutually repressive lineage-specific transcription factors A and B - enables two phenotypes from a common progenitor: (high A, low B) and (low A, high B). However, the dynamics of networks enabling differentiation of more than two phenotypes from a progenitor cell has not been well-studied. Here, we investigate the dynamics of a four-node network A, B, C, and D inhibiting each other, forming a toggle tetrahedron. Our simulations show that this network is multistable and predominantly allows for the co-existence of six hybrid phenotypes where two of the nodes are expressed relatively high as compared to the remaining two, for instance (high A, high B, low C, low D). Finally, we apply our results to understand naïve CD4<sup>+</sup> T cell differentiation into Th1, Th2, Th17 and Treg subsets, suggesting Th1/Th2/Th17/Treg decision-making to be a two-step process.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"123"},"PeriodicalIF":3.5,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11502801/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504871","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genome-scale metabolic models (GEMs) cover the entire list of metabolic genes in an organism and associated reactions, in a tissue/condition non-specific manner. RNA-seq provides crucial information to make the GEMs condition-specific. Integrative Metabolic Analysis Tool (iMAT) and Integrative Network Inference for Tissues (INIT) are the two most popular algorithms to create condition-specific GEMs from human transcriptome data. The normalization method of choice for raw RNA-seq count data affects the model content produced by these algorithms and their predictive accuracy. However, a benchmark of the RNA-seq normalization methods on the performance of iMAT and INIT algorithms is missing in the literature. Another important phenomenon is covariates such as age and gender in a dataset, and they can affect the predictivity of analysis. In this study, we aimed to compare five different RNA-seq data normalization methods (TPM, FPKM, TMM, GeTMM, and RLE) and covariate adjusted versions of the normalized data by mapping them on a human GEM using the iMAT and INIT algorithms to generate personalized metabolic models. We used RNA-seq data for Alzheimer's disease (AD) and lung adenocarcinoma (LUAD) patients. The results demonstrated that RNA-seq data normalized by the RLE, TMM, or GeTMM methods enabled the production of condition-specific metabolic models with considerably low variability in terms of the number of active reactions compared to the within-sample normalization methods (FPKM, TPM). Using these models, we could more accurately capture the disease-associated genes (average accuracy of ~0.80 for AD and ~0.67 for LUAD) for the RLE, TMM, and GeTMM normalization methods. An increase in the accuracies was observed for all the methods when covariate adjustment was applied. We found a similar accuracy trend when we compared the metabolites of perturbed reactions to metabolome data for AD. Together, our benchmark study shows that the between-sample RNA-seq normalization methods reduce false positive predictions at the expense of missing some true positive genes when mapped on GEMs.
{"title":"A benchmark of RNA-seq data normalization methods for transcriptome mapping on human genome-scale metabolic networks.","authors":"Hatice Büşra Lüleci, Dilara Uzuner, Müberra Fatma Cesur, Atılay İlgün, Elif Düz, Ecehan Abdik, Regan Odongo, Tunahan Çakır","doi":"10.1038/s41540-024-00448-z","DOIUrl":"https://doi.org/10.1038/s41540-024-00448-z","url":null,"abstract":"<p><p>Genome-scale metabolic models (GEMs) cover the entire list of metabolic genes in an organism and associated reactions, in a tissue/condition non-specific manner. RNA-seq provides crucial information to make the GEMs condition-specific. Integrative Metabolic Analysis Tool (iMAT) and Integrative Network Inference for Tissues (INIT) are the two most popular algorithms to create condition-specific GEMs from human transcriptome data. The normalization method of choice for raw RNA-seq count data affects the model content produced by these algorithms and their predictive accuracy. However, a benchmark of the RNA-seq normalization methods on the performance of iMAT and INIT algorithms is missing in the literature. Another important phenomenon is covariates such as age and gender in a dataset, and they can affect the predictivity of analysis. In this study, we aimed to compare five different RNA-seq data normalization methods (TPM, FPKM, TMM, GeTMM, and RLE) and covariate adjusted versions of the normalized data by mapping them on a human GEM using the iMAT and INIT algorithms to generate personalized metabolic models. We used RNA-seq data for Alzheimer's disease (AD) and lung adenocarcinoma (LUAD) patients. The results demonstrated that RNA-seq data normalized by the RLE, TMM, or GeTMM methods enabled the production of condition-specific metabolic models with considerably low variability in terms of the number of active reactions compared to the within-sample normalization methods (FPKM, TPM). Using these models, we could more accurately capture the disease-associated genes (average accuracy of ~0.80 for AD and ~0.67 for LUAD) for the RLE, TMM, and GeTMM normalization methods. An increase in the accuracies was observed for all the methods when covariate adjustment was applied. We found a similar accuracy trend when we compared the metabolites of perturbed reactions to metabolome data for AD. Together, our benchmark study shows that the between-sample RNA-seq normalization methods reduce false positive predictions at the expense of missing some true positive genes when mapped on GEMs.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"124"},"PeriodicalIF":3.5,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11502818/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1038/s41540-024-00453-2
Martina Conte, Vanesa Cabeza Fernández, F Javier Oliver, Tomás Alarcón, Juan Soler
Tumor hypoxia is a dynamic phenomenon marked by fluctuations in oxygen levels across both rapid (seconds to minutes) and slow (hours to days) time scales. While short hypoxia cycles are relatively well understood, the mechanisms behind longer cycles remain largely unclear. In this paper, we present a novel mechanistic mathematical model that explains slow hypoxia cycles through feedback loops involving vascular expansion and regression, oxygen-regulated tumor growth, and toxic cytokine production. Our study reveals that, for the emergence of slow hypoxia cycles, endothelial cells must adapt by decreasing receptor activation as ligand concentration increases. Additionally, the interaction between tumor cells and toxic cytokines influences frequency and intensity of these cycles. By examining the effects of pharmacological interventions, specifically poly (ADP-ribose) polymerase inhibitors, we also demonstrate how targeting cell proliferation can help regulate oxygen levels. Our findings enhance the understanding of hypoxia regulation and suggest PARP proteins as promising therapeutic targets.
{"title":"Emergence of cyclic hypoxia and the impact of PARP inhibitors on tumor progression.","authors":"Martina Conte, Vanesa Cabeza Fernández, F Javier Oliver, Tomás Alarcón, Juan Soler","doi":"10.1038/s41540-024-00453-2","DOIUrl":"10.1038/s41540-024-00453-2","url":null,"abstract":"<p><p>Tumor hypoxia is a dynamic phenomenon marked by fluctuations in oxygen levels across both rapid (seconds to minutes) and slow (hours to days) time scales. While short hypoxia cycles are relatively well understood, the mechanisms behind longer cycles remain largely unclear. In this paper, we present a novel mechanistic mathematical model that explains slow hypoxia cycles through feedback loops involving vascular expansion and regression, oxygen-regulated tumor growth, and toxic cytokine production. Our study reveals that, for the emergence of slow hypoxia cycles, endothelial cells must adapt by decreasing receptor activation as ligand concentration increases. Additionally, the interaction between tumor cells and toxic cytokines influences frequency and intensity of these cycles. By examining the effects of pharmacological interventions, specifically poly (ADP-ribose) polymerase inhibitors, we also demonstrate how targeting cell proliferation can help regulate oxygen levels. Our findings enhance the understanding of hypoxia regulation and suggest PARP proteins as promising therapeutic targets.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":"10 1","pages":"122"},"PeriodicalIF":3.5,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11494044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142471106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}