首页 > 最新文献

NPJ Systems Biology and Applications最新文献

英文 中文
Integrated-omics analysis with explainable deep networks on pathobiology of infant bronchiolitis. 利用可解释深度网络对婴儿支气管炎病理生物学进行综合组学分析
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-22 DOI: 10.1038/s41540-024-00420-x
Tadao Ooka, Naoto Usuyama, Ryohei Shibata, Michihito Kyo, Jonathan M Mansbach, Zhaozhong Zhu, Carlos A Camargo, Kohei Hasegawa

Bronchiolitis is the leading cause of infant hospitalization. However, the molecular networks driving bronchiolitis pathobiology remain unknown. Integrative molecular networks, including the transcriptome and metabolome, can identify functional and regulatory pathways contributing to disease severity. Here, we integrated nasopharyngeal transcriptome and metabolome data of 397 infants hospitalized with bronchiolitis in a 17-center prospective cohort study. Using an explainable deep network model, we identified an omics-cluster comprising 401 transcripts and 38 metabolites that distinguishes bronchiolitis severity (test-set AUC, 0.828). This omics-cluster derived a molecular network, where innate immunity-related metabolites (e.g., ceramides) centralized and were characterized by toll-like receptor (TLR) and NF-κB signaling pathways (both FDR < 0.001). The network analyses identified eight modules and 50 existing drug candidates for repurposing, including prostaglandin I2 analogs (e.g., iloprost), which promote anti-inflammatory effects through TLR signaling. Our approach facilitates not only the identification of molecular networks underlying infant bronchiolitis but the development of pioneering treatment strategies.

支气管炎是婴儿住院治疗的主要原因。然而,驱动支气管炎病理生物学的分子网络仍然未知。包括转录组和代谢组在内的整合分子网络可以确定导致疾病严重程度的功能和调控途径。在此,我们整合了一项 17 个中心的前瞻性队列研究中 397 名因支气管炎住院的婴儿的鼻咽转录组和代谢组数据。利用可解释的深度网络模型,我们发现了一个由 401 个转录本和 38 个代谢物组成的全局集群(omics-cluster),它能区分支气管炎的严重程度(测试集 AUC 为 0.828)。这个全元素集群衍生出一个分子网络,在这个网络中,先天免疫相关代谢物(如神经酰胺)处于中心位置,并以收费样受体(TLR)和 NF-κB 信号通路(均为 FDR 2 类似物(如伊洛前列素),通过 TLR 信号促进抗炎作用)为特征。我们的方法不仅有助于确定婴儿支气管炎的分子网络,还有助于开发开创性的治疗策略。
{"title":"Integrated-omics analysis with explainable deep networks on pathobiology of infant bronchiolitis.","authors":"Tadao Ooka, Naoto Usuyama, Ryohei Shibata, Michihito Kyo, Jonathan M Mansbach, Zhaozhong Zhu, Carlos A Camargo, Kohei Hasegawa","doi":"10.1038/s41540-024-00420-x","DOIUrl":"10.1038/s41540-024-00420-x","url":null,"abstract":"<p><p>Bronchiolitis is the leading cause of infant hospitalization. However, the molecular networks driving bronchiolitis pathobiology remain unknown. Integrative molecular networks, including the transcriptome and metabolome, can identify functional and regulatory pathways contributing to disease severity. Here, we integrated nasopharyngeal transcriptome and metabolome data of 397 infants hospitalized with bronchiolitis in a 17-center prospective cohort study. Using an explainable deep network model, we identified an omics-cluster comprising 401 transcripts and 38 metabolites that distinguishes bronchiolitis severity (test-set AUC, 0.828). This omics-cluster derived a molecular network, where innate immunity-related metabolites (e.g., ceramides) centralized and were characterized by toll-like receptor (TLR) and NF-κB signaling pathways (both FDR < 0.001). The network analyses identified eight modules and 50 existing drug candidates for repurposing, including prostaglandin I<sub>2</sub> analogs (e.g., iloprost), which promote anti-inflammatory effects through TLR signaling. Our approach facilitates not only the identification of molecular networks underlying infant bronchiolitis but the development of pioneering treatment strategies.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11341550/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142036502","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}
引用次数: 0
Assessing structural uncertainty of biochemical regulatory networks in metabolic pathways under varying data quality. 评估不同数据质量下代谢途径中生化调控网络结构的不确定性。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-22 DOI: 10.1038/s41540-024-00412-x
Yue Han, Mark P Styczynski

Ordinary differential equation (ODE) models are powerful tools for studying the dynamics of metabolic pathways. However, key challenges lie in constructing ODE models for metabolic pathways, specifically in our limited knowledge about which metabolite levels control which reaction rates. Identification of these regulatory networks is further complicated by the limited availability of relevant data. Here, we assess the conditions under which it is feasible to accurately identify regulatory networks in metabolic pathways by computationally fitting candidate network models with biochemical systems theory (BST) kinetics to data of varying quality. We use network motifs commonly found in metabolic pathways as a simplified testbed. Key features correlated with the level of difficulty in identifying the correct regulatory network were identified, highlighting the impact of sampling rate, data noise, and data incompleteness on structural uncertainty. We found that for a simple branched network motif with an equal number of metabolites and fluxes, identification of the correct regulatory network can be largely achieved and is robust to missing one of the metabolite profiles. However, with a bi-substrate bi-product reaction or more fluxes than metabolites in the network motif, the identification becomes more challenging. Stronger regulatory interactions and higher metabolite concentrations were found to be correlated with less structural uncertainty. These results could aid efforts to predict whether the true metabolic regulatory network can be computationally identified for a given stoichiometric network topology and dataset quality, thus helping to identify optimal measures to mitigate such identifiability issues in kinetic model development.

常微分方程(ODE)模型是研究代谢途径动态的有力工具。然而,为代谢途径构建 ODE 模型的关键挑战在于我们对哪些代谢物水平控制哪些反应速率的了解有限。由于相关数据有限,这些调控网络的识别变得更加复杂。在这里,我们通过计算将候选网络模型与生化系统理论(BST)动力学拟合到不同质量的数据中,来评估在什么条件下可以准确识别代谢途径中的调控网络。我们使用代谢途径中常见的网络图案作为简化的试验平台。我们确定了与识别正确调控网络的难度相关的关键特征,突出了采样率、数据噪声和数据不完整性对结构不确定性的影响。我们发现,对于代谢物和通量数量相等的简单分支网络图案,基本上可以识别出正确的调控网络,而且对缺少其中一个代谢物图谱的情况也很稳健。然而,当网络图案中存在双底物双产物反应或通量多于代谢物时,识别工作就变得更具挑战性。研究发现,较强的调控相互作用和较高的代谢物浓度与较小的结构不确定性相关。这些结果有助于预测在给定的化学计量网络拓扑结构和数据集质量下,是否能通过计算识别出真正的代谢调控网络,从而帮助确定最佳措施,以减轻动力学模型开发中的可识别性问题。
{"title":"Assessing structural uncertainty of biochemical regulatory networks in metabolic pathways under varying data quality.","authors":"Yue Han, Mark P Styczynski","doi":"10.1038/s41540-024-00412-x","DOIUrl":"10.1038/s41540-024-00412-x","url":null,"abstract":"<p><p>Ordinary differential equation (ODE) models are powerful tools for studying the dynamics of metabolic pathways. However, key challenges lie in constructing ODE models for metabolic pathways, specifically in our limited knowledge about which metabolite levels control which reaction rates. Identification of these regulatory networks is further complicated by the limited availability of relevant data. Here, we assess the conditions under which it is feasible to accurately identify regulatory networks in metabolic pathways by computationally fitting candidate network models with biochemical systems theory (BST) kinetics to data of varying quality. We use network motifs commonly found in metabolic pathways as a simplified testbed. Key features correlated with the level of difficulty in identifying the correct regulatory network were identified, highlighting the impact of sampling rate, data noise, and data incompleteness on structural uncertainty. We found that for a simple branched network motif with an equal number of metabolites and fluxes, identification of the correct regulatory network can be largely achieved and is robust to missing one of the metabolite profiles. However, with a bi-substrate bi-product reaction or more fluxes than metabolites in the network motif, the identification becomes more challenging. Stronger regulatory interactions and higher metabolite concentrations were found to be correlated with less structural uncertainty. These results could aid efforts to predict whether the true metabolic regulatory network can be computationally identified for a given stoichiometric network topology and dataset quality, thus helping to identify optimal measures to mitigate such identifiability issues in kinetic model development.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11341918/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142036501","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}
引用次数: 0
Using DeepSignalingFlow to mine signaling flows interpreting mechanism of synergy of cocktails. 使用 DeepSignalingFlow 挖掘信号流,解释鸡尾酒的协同作用机制。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-21 DOI: 10.1038/s41540-024-00421-w
Heming Zhang, Yixin Chen, Philip Payne, Fuhai Li

Complex signaling pathways are believed to be responsible for drug resistance. Drug combinations perturbing multiple signaling targets have the potential to reduce drug resistance. The large-scale multi-omic datasets and experimental drug combination synergistic score data are valuable resources to study mechanisms of synergy (MoS) to guide the development of precision drug combinations. However, signaling patterns of MoS are complex and remain unclear, and thus it is challenging to identify synergistic drug combinations in clinical. Herein, we proposed a novel integrative and interpretable graph AI model, DeepSignalingFlow, to uncover the MoS by integrating and mining multi-omic data. The major innovation is that we uncover MoS by modeling the signaling flow from multi-omic features of essential disease proteins to the drug targets, which has not been introduced by the existing models. The model performance was assessed utilizing four distinct drug combination synergy evaluation datasets, i.e., NCI ALMANAC, O'Neil, DrugComb, and DrugCombDB. The comparison results showed that the proposed model outperformed existing graph AI models in terms of synergy score prediction, and can interpret MoS using the core signaling flows. The code is publicly accessible via Github: https://github.com/FuhaiLiAiLab/DeepSignalingFlow.

复杂的信号通路被认为是导致耐药性的原因。干扰多个信号靶点的药物组合有可能减少耐药性。大规模多组学数据集和实验性药物组合协同得分数据是研究协同机制(MoS)的宝贵资源,可用于指导精准药物组合的开发。然而,MoS 的信号转导模式十分复杂,目前仍不清楚,因此在临床上识别协同药物组合具有挑战性。在此,我们提出了一种新颖的整合性和可解释性图人工智能模型--DeepSignalingFlow,通过整合和挖掘多组学数据来揭示MoS。该模型的主要创新之处在于,我们通过模拟从基本疾病蛋白的多组学特征到药物靶点的信号流来揭示MoS,而现有模型并未引入这种信号流。我们利用 NCI ALMANAC、O'Neil、DrugComb 和 DrugCombDB 这四个不同的药物组合协同作用评估数据集对该模型的性能进行了评估。比较结果表明,所提出的模型在协同作用得分预测方面优于现有的图人工智能模型,并能利用核心信号流解释 MoS。代码可通过 Github 公开访问:https://github.com/FuhaiLiAiLab/DeepSignalingFlow。
{"title":"Using DeepSignalingFlow to mine signaling flows interpreting mechanism of synergy of cocktails.","authors":"Heming Zhang, Yixin Chen, Philip Payne, Fuhai Li","doi":"10.1038/s41540-024-00421-w","DOIUrl":"10.1038/s41540-024-00421-w","url":null,"abstract":"<p><p>Complex signaling pathways are believed to be responsible for drug resistance. Drug combinations perturbing multiple signaling targets have the potential to reduce drug resistance. The large-scale multi-omic datasets and experimental drug combination synergistic score data are valuable resources to study mechanisms of synergy (MoS) to guide the development of precision drug combinations. However, signaling patterns of MoS are complex and remain unclear, and thus it is challenging to identify synergistic drug combinations in clinical. Herein, we proposed a novel integrative and interpretable graph AI model, DeepSignalingFlow, to uncover the MoS by integrating and mining multi-omic data. The major innovation is that we uncover MoS by modeling the signaling flow from multi-omic features of essential disease proteins to the drug targets, which has not been introduced by the existing models. The model performance was assessed utilizing four distinct drug combination synergy evaluation datasets, i.e., NCI ALMANAC, O'Neil, DrugComb, and DrugCombDB. The comparison results showed that the proposed model outperformed existing graph AI models in terms of synergy score prediction, and can interpret MoS using the core signaling flows. The code is publicly accessible via Github: https://github.com/FuhaiLiAiLab/DeepSignalingFlow.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11339460/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142018176","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}
引用次数: 0
Spatial computational modelling illuminates the role of the tumour microenvironment for treating glioblastoma with immunotherapies. 空间计算模型揭示了肿瘤微环境在利用免疫疗法治疗胶质母细胞瘤中的作用。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-18 DOI: 10.1038/s41540-024-00419-4
Blanche Mongeon, Julien Hébert-Doutreloux, Anudeep Surendran, Elham Karimi, Benoit Fiset, Daniela F Quail, Logan A Walsh, Adrianne L Jenner, Morgan Craig

Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.

胶质母细胞瘤是成人中最常见、最致命的脑肿瘤,在目前的治疗标准下,中位生存期为 15 个月。为了改善这一终点,人们对免疫检查点抑制剂和溶瘤病毒等免疫疗法进行了广泛研究。然而,迄今为止,大多数研究都以失败告终。为了提高免疫疗法治疗胶质母细胞瘤的疗效,可以利用成像质控细胞仪等新型单细胞成像模式,并将其与计算模型相结合。这样就能更好地了解肿瘤微环境及其对这种难以治疗的肿瘤的治疗成败所起的作用。在这里,我们实施了一个基于代理的模型,该模型可对联合化疗、溶瘤病毒和免疫检查点抑制剂治疗胶质母细胞瘤进行空间预测。我们利用患者成像质谱数据对模型进行了初始化,以预测患者的特异性反应,并发现溶瘤病毒会驱动由瘤内细胞密度决定的联合治疗反应。我们发现,肿瘤细胞密度较高的肿瘤对治疗的反应更好。在固定癌细胞数量的情况下,治疗效果与 CD4 + T 细胞有关,其次与巨噬细胞数量有关。重要的是,我们的模拟结果表明,必须小心整合空间数据和基于代理的模型,才能有效捕捉肿瘤内的动态变化。总之,这项研究强调了使用预测性空间建模来更好地理解癌症免疫疗法的治疗动态,同时突出了在模型设计和实施过程中需要考虑的关键因素。
{"title":"Spatial computational modelling illuminates the role of the tumour microenvironment for treating glioblastoma with immunotherapies.","authors":"Blanche Mongeon, Julien Hébert-Doutreloux, Anudeep Surendran, Elham Karimi, Benoit Fiset, Daniela F Quail, Logan A Walsh, Adrianne L Jenner, Morgan Craig","doi":"10.1038/s41540-024-00419-4","DOIUrl":"10.1038/s41540-024-00419-4","url":null,"abstract":"<p><p>Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11330976/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142000470","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}
引用次数: 0
Insights gained from computational modeling of YAP/TAZ signaling for cellular mechanotransduction. 从细胞机械传导的 YAP/TAZ 信号的计算建模中获得的启示。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-15 DOI: 10.1038/s41540-024-00414-9
Hamidreza Jafarinia, Ali Khalilimeybodi, Jorge Barrasa-Fano, Stephanie I Fraley, Padmini Rangamani, Aurélie Carlier

YAP/TAZ signaling pathway is regulated by a multiplicity of feedback loops, crosstalk with other pathways, and both mechanical and biochemical stimuli. Computational modeling serves as a powerful tool to unravel how these different factors can regulate YAP/TAZ, emphasizing biophysical modeling as an indispensable tool for deciphering mechanotransduction and its regulation of cell fate. We provide a critical review of the current state-of-the-art of computational models focused on YAP/TAZ signaling.

YAP/TAZ信号通路受多种反馈回路、与其他通路的串扰以及机械和生化刺激的调控。计算建模是揭示这些不同因素如何调控 YAP/TAZ 的有力工具,强调生物物理建模是解密机械传导及其对细胞命运调控不可或缺的工具。我们对当前以 YAP/TAZ 信号转导为重点的计算模型的最新进展进行了深入评述。
{"title":"Insights gained from computational modeling of YAP/TAZ signaling for cellular mechanotransduction.","authors":"Hamidreza Jafarinia, Ali Khalilimeybodi, Jorge Barrasa-Fano, Stephanie I Fraley, Padmini Rangamani, Aurélie Carlier","doi":"10.1038/s41540-024-00414-9","DOIUrl":"10.1038/s41540-024-00414-9","url":null,"abstract":"<p><p>YAP/TAZ signaling pathway is regulated by a multiplicity of feedback loops, crosstalk with other pathways, and both mechanical and biochemical stimuli. Computational modeling serves as a powerful tool to unravel how these different factors can regulate YAP/TAZ, emphasizing biophysical modeling as an indispensable tool for deciphering mechanotransduction and its regulation of cell fate. We provide a critical review of the current state-of-the-art of computational models focused on YAP/TAZ signaling.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11327324/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141988476","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}
引用次数: 0
Practical parameter identifiability and handling of censored data with Bayesian inference in mathematical tumour models. 在肿瘤数学模型中使用贝叶斯推断法进行实际参数可识别性和删减数据处理。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-14 DOI: 10.1038/s41540-024-00409-6
Jamie Porthiyas, Daniel Nussey, Catherine A A Beauchemin, Donald C Warren, Christian Quirouette, Kathleen P Wilkie

Mechanistic mathematical models (MMs) are a powerful tool to help us understand and predict the dynamics of tumour growth under various conditions. In this work, we use 5 MMs with an increasing number of parameters to explore how certain (often overlooked) decisions in estimating parameters from data of experimental tumour growth affect the outcome of the analysis. In particular, we propose a framework for including tumour volume measurements that fall outside the upper and lower limits of detection, which are normally discarded. We demonstrate how excluding censored data results in an overestimation of the initial tumour volume and the MM-predicted tumour volumes prior to the first measurements, and an underestimation of the carrying capacity and the MM-predicted tumour volumes beyond the latest measurable time points. We show in which way the choice of prior for the MM parameters can impact the posterior distributions, and illustrate that reporting the most likely parameters and their 95% credible interval can lead to confusing or misleading interpretations. We hope this work will encourage others to carefully consider choices made in parameter estimation and to adopt the approaches we put forward herein.

机理数学模型(MMs)是帮助我们理解和预测各种条件下肿瘤生长动态的有力工具。在这项工作中,我们使用了参数数量不断增加的 5 个 MM,来探讨从肿瘤生长实验数据中估算参数时的某些(通常被忽视的)决定是如何影响分析结果的。特别是,我们提出了一个框架,用于将通常被剔除的、超出检测上下限的肿瘤体积测量数据包括在内。我们展示了排除删减数据如何导致高估首次测量前的初始肿瘤体积和 MM 预测肿瘤体积,以及低估承载能力和超过最新可测量时间点的 MM 预测肿瘤体积。我们展示了 MM 参数先验值的选择会以何种方式影响后验分布,并说明报告最可能的参数及其 95% 可信区间可能会导致混乱或误导性解释。我们希望这项工作能鼓励其他人仔细考虑参数估计中的选择,并采用我们在此提出的方法。
{"title":"Practical parameter identifiability and handling of censored data with Bayesian inference in mathematical tumour models.","authors":"Jamie Porthiyas, Daniel Nussey, Catherine A A Beauchemin, Donald C Warren, Christian Quirouette, Kathleen P Wilkie","doi":"10.1038/s41540-024-00409-6","DOIUrl":"10.1038/s41540-024-00409-6","url":null,"abstract":"<p><p>Mechanistic mathematical models (MMs) are a powerful tool to help us understand and predict the dynamics of tumour growth under various conditions. In this work, we use 5 MMs with an increasing number of parameters to explore how certain (often overlooked) decisions in estimating parameters from data of experimental tumour growth affect the outcome of the analysis. In particular, we propose a framework for including tumour volume measurements that fall outside the upper and lower limits of detection, which are normally discarded. We demonstrate how excluding censored data results in an overestimation of the initial tumour volume and the MM-predicted tumour volumes prior to the first measurements, and an underestimation of the carrying capacity and the MM-predicted tumour volumes beyond the latest measurable time points. We show in which way the choice of prior for the MM parameters can impact the posterior distributions, and illustrate that reporting the most likely parameters and their 95% credible interval can lead to confusing or misleading interpretations. We hope this work will encourage others to carefully consider choices made in parameter estimation and to adopt the approaches we put forward herein.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11324876/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141982878","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}
引用次数: 0
Hybridizing mechanistic modeling and deep learning for personalized survival prediction after immune checkpoint inhibitor immunotherapy. 杂交机理建模和深度学习,用于免疫检查点抑制剂免疫疗法后的个性化生存预测。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-14 DOI: 10.1038/s41540-024-00415-8
Joseph D Butner, Prashant Dogra, Caroline Chung, Eugene J Koay, James W Welsh, David S Hong, Vittorio Cristini, Zhihui Wang

We present a study where predictive mechanistic modeling is combined with deep learning methods to predict individual patient survival probabilities under immune checkpoint inhibitor (ICI) immunotherapy. This hybrid approach enables prediction based on both measures that are calculable from mechanistic models of key mechanisms underlying ICI therapy that may not be directly measurable in the clinic and easily measurable quantities or patient characteristics that are not always readily incorporated into predictive mechanistic models. A deep learning time-to-event predictive model trained on a hybrid mechanistic + clinical data set from 93 patients achieved higher per-patient predictive accuracy based on event-time concordance, Brier score, and negative binomial log-likelihood-based criteria than when trained on only mechanistic model-derived values or only clinical data. Feature importance analysis revealed that both clinical and model-derived parameters play prominent roles in increasing prediction accuracy, further supporting the advantage of our hybrid approach.

我们介绍了一项研究,该研究将预测性机理建模与深度学习方法相结合,预测免疫检查点抑制剂(ICI)免疫疗法下个体患者的生存概率。这种混合方法既能根据 ICI 治疗关键机制的机理模型计算出的指标(这些指标在临床上可能无法直接测量)进行预测,又能根据易于测量的数量或患者特征进行预测,而这些数量或特征并不总是很容易纳入预测性机理模型中。基于事件时间一致性、布赖尔评分和基于负二叉对数似然法的标准,在来自 93 名患者的机理+临床混合数据集上训练的深度学习时间到事件预测模型比仅在机理模型衍生值或仅在临床数据上训练的模型获得了更高的单个患者预测准确率。特征重要性分析表明,临床参数和模型衍生参数在提高预测准确率方面都发挥了重要作用,这进一步证明了我们的混合方法的优势。
{"title":"Hybridizing mechanistic modeling and deep learning for personalized survival prediction after immune checkpoint inhibitor immunotherapy.","authors":"Joseph D Butner, Prashant Dogra, Caroline Chung, Eugene J Koay, James W Welsh, David S Hong, Vittorio Cristini, Zhihui Wang","doi":"10.1038/s41540-024-00415-8","DOIUrl":"10.1038/s41540-024-00415-8","url":null,"abstract":"<p><p>We present a study where predictive mechanistic modeling is combined with deep learning methods to predict individual patient survival probabilities under immune checkpoint inhibitor (ICI) immunotherapy. This hybrid approach enables prediction based on both measures that are calculable from mechanistic models of key mechanisms underlying ICI therapy that may not be directly measurable in the clinic and easily measurable quantities or patient characteristics that are not always readily incorporated into predictive mechanistic models. A deep learning time-to-event predictive model trained on a hybrid mechanistic + clinical data set from 93 patients achieved higher per-patient predictive accuracy based on event-time concordance, Brier score, and negative binomial log-likelihood-based criteria than when trained on only mechanistic model-derived values or only clinical data. Feature importance analysis revealed that both clinical and model-derived parameters play prominent roles in increasing prediction accuracy, further supporting the advantage of our hybrid approach.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11324794/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141982877","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}
引用次数: 0
A practically efficient algorithm for identifying critical control proteins in directed probabilistic biological networks 在有向概率生物网络中识别关键控制蛋白的实用高效算法
IF 4 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-12 DOI: 10.1038/s41540-024-00411-y
Yusuke Tokuhara, Tatsuya Akutsu, Jean-Marc Schwartz, Jose C. Nacher

Network controllability is unifying the traditional control theory with the structural network information rooted in many large-scale biological systems of interest, from intracellular networks in molecular biology to brain neuronal networks. In controllability approaches, the set of minimum driver nodes is not unique, and critical nodes are the most important control elements because they appear in all possible solution sets. On the other hand, a common but largely unexplored feature in network control approaches is the probabilistic failure of edges or the uncertainty in the determination of interactions between molecules. This is particularly true when directed probabilistic interactions are considered. Until now, no efficient algorithm existed to determine critical nodes in probabilistic directed networks. Here we present a probabilistic control model based on a minimum dominating set framework that integrates the probabilistic nature of directed edges between molecules and determines the critical control nodes that drive the entire network functionality. The proposed algorithm, combined with the developed mathematical tools, offers practical efficiency in determining critical control nodes in large probabilistic networks. The method is then applied to the human intracellular signal transduction network revealing that critical control nodes are associated with important biological features and perturbed sets of genes in human diseases, including SARS-CoV-2 target proteins and rare disorders. We believe that the proposed methodology can be useful to investigate multiple biological systems in which directed edges are probabilistic in nature, both in natural systems or when determined with large uncertainties in-silico.

从分子生物学的细胞内网络到大脑神经元网络,网络可控性是传统控制理论与结构网络信息的统一。在可控性方法中,最小驱动节点集并不是唯一的,关键节点是最重要的控制元素,因为它们出现在所有可能的解集中。另一方面,在网络控制方法中,一个常见但基本未被探索的特征是边的概率失效或分子间相互作用的确定存在不确定性。在考虑有向概率相互作用时尤其如此。到目前为止,还没有一种有效的算法来确定概率有向网络中的关键节点。在这里,我们提出了一种基于最小支配集框架的概率控制模型,它整合了分子间有向边缘的概率性质,并确定了驱动整个网络功能的关键控制节点。所提出的算法与所开发的数学工具相结合,在确定大型概率网络的关键控制节点方面具有实际效率。我们将该方法应用于人类细胞内信号转导网络,发现关键控制节点与人类疾病(包括 SARS-CoV-2 目标蛋白和罕见疾病)中的重要生物学特征和受干扰基因集相关。我们相信,无论是在自然系统中,还是在具有较大不确定性的实验室测定中,所提出的方法都可用于研究有向边缘具有概率性质的多种生物系统。
{"title":"A practically efficient algorithm for identifying critical control proteins in directed probabilistic biological networks","authors":"Yusuke Tokuhara, Tatsuya Akutsu, Jean-Marc Schwartz, Jose C. Nacher","doi":"10.1038/s41540-024-00411-y","DOIUrl":"https://doi.org/10.1038/s41540-024-00411-y","url":null,"abstract":"<p>Network controllability is unifying the traditional control theory with the structural network information rooted in many large-scale biological systems of interest, from intracellular networks in molecular biology to brain neuronal networks. In controllability approaches, the set of minimum driver nodes is not unique, and critical nodes are the most important control elements because they appear in all possible solution sets. On the other hand, a common but largely unexplored feature in network control approaches is the probabilistic failure of edges or the uncertainty in the determination of interactions between molecules. This is particularly true when directed probabilistic interactions are considered. Until now, no efficient algorithm existed to determine critical nodes in probabilistic directed networks. Here we present a probabilistic control model based on a minimum dominating set framework that integrates the probabilistic nature of directed edges between molecules and determines the critical control nodes that drive the entire network functionality. The proposed algorithm, combined with the developed mathematical tools, offers practical efficiency in determining critical control nodes in large probabilistic networks. The method is then applied to the human intracellular signal transduction network revealing that critical control nodes are associated with important biological features and perturbed sets of genes in human diseases, including SARS-CoV-2 target proteins and rare disorders. We believe that the proposed methodology can be useful to investigate multiple biological systems in which directed edges are probabilistic in nature, both in natural systems or when determined with large uncertainties in-silico.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141968650","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transient frequency preference responses in cell signaling systems. 细胞信号系统中的瞬态频率偏好反应。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-11 DOI: 10.1038/s41540-024-00413-w
Candela L Szischik, Juliana Reves Szemere, Rocío Balderrama, Constanza Sánchez de la Vega, Alejandra C Ventura

Ligand-receptor systems, covalent modification cycles, and transcriptional networks are the fundamental components of cell signaling and gene expression systems. While their behavior in reaching a steady-state regime under step-like stimulation is well understood, their response under repetitive stimulation, particularly at early time stages is poorly characterized. Yet, early-stage responses to external inputs are arguably as informative as late-stage ones. In simple systems, a periodic stimulation elicits an initial transient response, followed by periodic behavior. Transient responses are relevant when the stimulation has a limited time span, or when the stimulated component's timescale is slow as compared to the timescales of the downstream processes, in which case the latter processes may be capturing only those transients. In this study, we analyze the frequency response of simple motifs at different time stages. We use dose-conserved pulsatile input signals and consider different metrics versus frequency curves. We show that in ligand-receptor systems, there is a frequency preference response in some specific metrics during the transient stages, which is not present in the periodic regime. We suggest this is a general system-level mechanism that cells may use to filter input signals that have consequences for higher order circuits. In addition, we evaluate how the described behavior in isolated motifs is reflected in similar types of responses in cascades and pathways of which they are a part. Our studies suggest that transient frequency preferences are important dynamic features of cell signaling and gene expression systems, which have been overlooked.

配体-受体系统、共价修饰循环和转录网络是细胞信号和基因表达系统的基本组成部分。虽然人们对它们在阶跃式刺激下达到稳态的行为非常了解,但它们在重复性刺激下的反应,尤其是在早期阶段的反应,却鲜为人知。然而,早期对外部输入的反应可以说与晚期的反应一样具有参考价值。在简单的系统中,周期性刺激会引起最初的瞬态反应,随后出现周期性行为。当刺激的时间跨度有限,或受刺激成分的时间尺度与下游过程的时间尺度相比较慢时,瞬态反应就会出现,在这种情况下,下游过程可能只能捕捉到这些瞬态反应。在本研究中,我们分析了简单图案在不同时间阶段的频率响应。我们使用剂量保守的脉冲输入信号,并考虑了不同的指标与频率曲线。我们发现,在配体-受体系统中,某些特定指标在瞬态阶段存在频率偏好响应,而在周期机制中则不存在。我们认为这是一种一般的系统级机制,细胞可能会利用这种机制来过滤输入信号,从而对高阶电路产生影响。此外,我们还评估了孤立图案中描述的行为如何反映在级联和通路中类似类型的反应中,而它们是级联和通路的一部分。我们的研究表明,瞬态频率偏好是细胞信号传导和基因表达系统的重要动态特征,而这些特征一直被忽视。
{"title":"Transient frequency preference responses in cell signaling systems.","authors":"Candela L Szischik, Juliana Reves Szemere, Rocío Balderrama, Constanza Sánchez de la Vega, Alejandra C Ventura","doi":"10.1038/s41540-024-00413-w","DOIUrl":"10.1038/s41540-024-00413-w","url":null,"abstract":"<p><p>Ligand-receptor systems, covalent modification cycles, and transcriptional networks are the fundamental components of cell signaling and gene expression systems. While their behavior in reaching a steady-state regime under step-like stimulation is well understood, their response under repetitive stimulation, particularly at early time stages is poorly characterized. Yet, early-stage responses to external inputs are arguably as informative as late-stage ones. In simple systems, a periodic stimulation elicits an initial transient response, followed by periodic behavior. Transient responses are relevant when the stimulation has a limited time span, or when the stimulated component's timescale is slow as compared to the timescales of the downstream processes, in which case the latter processes may be capturing only those transients. In this study, we analyze the frequency response of simple motifs at different time stages. We use dose-conserved pulsatile input signals and consider different metrics versus frequency curves. We show that in ligand-receptor systems, there is a frequency preference response in some specific metrics during the transient stages, which is not present in the periodic regime. We suggest this is a general system-level mechanism that cells may use to filter input signals that have consequences for higher order circuits. In addition, we evaluate how the described behavior in isolated motifs is reflected in similar types of responses in cascades and pathways of which they are a part. Our studies suggest that transient frequency preferences are important dynamic features of cell signaling and gene expression systems, which have been overlooked.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11317535/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141917152","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}
引用次数: 0
High-affinity biomolecular interactions are modulated by low-affinity binders. 高亲和力生物分子相互作用受低亲和力粘合剂的调节。
IF 3.5 2区 生物学 Q1 MATHEMATICAL & COMPUTATIONAL BIOLOGY Pub Date : 2024-08-10 DOI: 10.1038/s41540-024-00410-z
S Mukundan, Girish Deshpande, M S Madhusudhan

The strength of molecular interactions is characterized by their dissociation constants (KD). Only high-affinity interactions (KD ≤ 10-8 M) are extensively investigated and support binary on/off switches. However, such analyses have discounted the presence of low-affinity binders (KD > 10-5 M) in the cellular environment. We assess the potential influence of low-affinity binders on high-affinity interactions. By employing Gillespie stochastic simulations and continuous methods, we demonstrate that the presence of low-affinity binders can alter the kinetics and the steady state of high-affinity interactions. We refer to this effect as 'herd regulation' and have evaluated its possible impact in two different contexts including sex determination in Drosophila melanogaster and in signalling systems that employ molecular thresholds. We have also suggested experiments to validate herd regulation in vitro. We speculate that low-affinity binders are prevalent in biological contexts where the outcomes depend on molecular thresholds impacting homoeostatic regulation.

分子相互作用的强度以其解离常数(KD)为特征。只有高亲和力的相互作用(KD ≤ 10-8 M)才被广泛研究,并支持二元开/关开关。然而,这些分析忽略了细胞环境中低亲和力粘合剂(KD > 10-5 M)的存在。我们评估了低亲和力结合剂对高亲和力相互作用的潜在影响。通过采用 Gillespie 随机模拟和连续方法,我们证明了低亲和力结合剂的存在会改变高亲和力相互作用的动力学和稳定状态。我们将这种效应称为 "群体调节",并评估了它在两种不同情况下可能产生的影响,包括黑腹果蝇的性别决定和采用分子阈值的信号系统。我们还提出了在体外验证群体调节的实验建议。我们推测,低亲和力结合剂普遍存在于生物环境中,其结果取决于影响同态调节的分子阈值。
{"title":"High-affinity biomolecular interactions are modulated by low-affinity binders.","authors":"S Mukundan, Girish Deshpande, M S Madhusudhan","doi":"10.1038/s41540-024-00410-z","DOIUrl":"10.1038/s41540-024-00410-z","url":null,"abstract":"<p><p>The strength of molecular interactions is characterized by their dissociation constants (K<sub>D</sub>). Only high-affinity interactions (K<sub>D</sub> ≤ 10<sup>-8</sup> M) are extensively investigated and support binary on/off switches. However, such analyses have discounted the presence of low-affinity binders (K<sub>D</sub> > 10<sup>-5</sup> M) in the cellular environment. We assess the potential influence of low-affinity binders on high-affinity interactions. By employing Gillespie stochastic simulations and continuous methods, we demonstrate that the presence of low-affinity binders can alter the kinetics and the steady state of high-affinity interactions. We refer to this effect as 'herd regulation' and have evaluated its possible impact in two different contexts including sex determination in Drosophila melanogaster and in signalling systems that employ molecular thresholds. We have also suggested experiments to validate herd regulation in vitro. We speculate that low-affinity binders are prevalent in biological contexts where the outcomes depend on molecular thresholds impacting homoeostatic regulation.</p>","PeriodicalId":19345,"journal":{"name":"NPJ Systems Biology and Applications","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11316740/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141913464","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}
引用次数: 0
期刊
NPJ Systems Biology and Applications
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1