首页 > 最新文献

Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing最新文献

英文 中文
MaTiLDA: An Integrated Machine Learning and Topological Data Analysis Platform for Brain Network Dynamics. MaTiLDA:用于脑网络动力学的机器学习和拓扑数据分析集成平台。
Katrina Prantzalos, Dipak Upadhyaya, Nassim Shafiabadi, Guadalupe Fernandez-BacaVaca, Nick Gurski, Kenneth Yoshimoto, Subhashini Sivagnanam, Amitava Majumdar, Satya S Sahoo

Topological data analysis (TDA) combined with machine learning (ML) algorithms is a powerful approach for investigating complex brain interaction patterns in neurological disorders such as epilepsy. However, the use of ML algorithms and TDA for analysis of aberrant brain interactions requires substantial domain knowledge in computing as well as pure mathematics. To lower the threshold for clinical and computational neuroscience researchers to effectively use ML algorithms together with TDA to study neurological disorders, we introduce an integrated web platform called MaTiLDA. MaTiLDA is the first tool that enables users to intuitively use TDA methods together with ML models to characterize interaction patterns derived from neurophysiological signal data such as electroencephalogram (EEG) recorded during routine clinical practice. MaTiLDA features support for TDA methods, such as persistent homology, that enable classification of signal data using ML models to provide insights into complex brain interaction patterns in neurological disorders. We demonstrate the practical use of MaTiLDA by analyzing high-resolution intracranial EEG from refractory epilepsy patients to characterize the distinct phases of seizure propagation to different brain regions. The MaTiLDA platform is available at: https://bmhinformatics.case.edu/nicworkflow/MaTiLDA.

拓扑数据分析(TDA)与机器学习(ML)算法相结合,是研究癫痫等神经系统疾病中复杂的大脑交互模式的有力方法。然而,使用 ML 算法和 TDA 分析异常大脑交互需要大量的计算领域知识和纯数学知识。为了降低临床和计算神经科学研究人员有效使用 ML 算法和 TDA 研究神经系统疾病的门槛,我们推出了一个名为 MaTiLDA 的集成网络平台。MaTiLDA 是第一个能让用户直观地使用 TDA 方法和 ML 模型来描述从神经生理学信号数据(如常规临床实践中记录的脑电图)中得出的交互模式的工具。MaTiLDA 支持持续同源性等 TDA 方法,可使用 ML 模型对信号数据进行分类,从而深入了解神经系统疾病中复杂的大脑交互模式。通过分析难治性癫痫患者的高分辨率颅内脑电图,我们展示了 MaTiLDA 的实际应用,以描述癫痫发作向不同脑区传播的不同阶段。MaTiLDA平台的网址是:https://bmhinformatics.case.edu/nicworkflow/MaTiLDA。
{"title":"MaTiLDA: An Integrated Machine Learning and Topological Data Analysis Platform for Brain Network Dynamics.","authors":"Katrina Prantzalos, Dipak Upadhyaya, Nassim Shafiabadi, Guadalupe Fernandez-BacaVaca, Nick Gurski, Kenneth Yoshimoto, Subhashini Sivagnanam, Amitava Majumdar, Satya S Sahoo","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Topological data analysis (TDA) combined with machine learning (ML) algorithms is a powerful approach for investigating complex brain interaction patterns in neurological disorders such as epilepsy. However, the use of ML algorithms and TDA for analysis of aberrant brain interactions requires substantial domain knowledge in computing as well as pure mathematics. To lower the threshold for clinical and computational neuroscience researchers to effectively use ML algorithms together with TDA to study neurological disorders, we introduce an integrated web platform called MaTiLDA. MaTiLDA is the first tool that enables users to intuitively use TDA methods together with ML models to characterize interaction patterns derived from neurophysiological signal data such as electroencephalogram (EEG) recorded during routine clinical practice. MaTiLDA features support for TDA methods, such as persistent homology, that enable classification of signal data using ML models to provide insights into complex brain interaction patterns in neurological disorders. We demonstrate the practical use of MaTiLDA by analyzing high-resolution intracranial EEG from refractory epilepsy patients to characterize the distinct phases of seizure propagation to different brain regions. The MaTiLDA platform is available at: https://bmhinformatics.case.edu/nicworkflow/MaTiLDA.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075190","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Session Introduction: Precision Medicine: Innovative methods for advanced understanding of molecular underpinnings of disease. 会议简介:精准医学:通过创新方法深入了解疾病的分子基础。
Yana Bromberg, Hannah Carter, Steven E Brenner

Precision medicine, also often referred to as personalized medicine, targets the development of treatments and preventative measures specific to the individual's genomic signatures, lifestyle, and environmental conditions. The series of Precision Medicine sessions in PSB has continuously highlighted the advances in this field. Our 2024 collection of manuscripts showcases algorithmic advances that integrate data from distinct modalities and introduce innovative approaches to extract new, medically relevant information from existing data. These evolving technology and analytical methods promise to bring closer the goals of precision medicine to improve health and increase lifespan.

精准医学,也常被称为个性化医学,其目标是针对个人的基因组特征、生活方式和环境条件,开发特定的治疗和预防措施。PSB的精准医学系列会议不断强调这一领域的进展。我们的2024年手稿集展示了算法的进步,这些算法整合了来自不同模式的数据,并引入创新方法从现有数据中提取新的医学相关信息。这些不断发展的技术和分析方法有望进一步实现精准医学的目标,改善健康状况,延长寿命。
{"title":"Session Introduction: Precision Medicine: Innovative methods for advanced understanding of molecular underpinnings of disease.","authors":"Yana Bromberg, Hannah Carter, Steven E Brenner","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Precision medicine, also often referred to as personalized medicine, targets the development of treatments and preventative measures specific to the individual's genomic signatures, lifestyle, and environmental conditions. The series of Precision Medicine sessions in PSB has continuously highlighted the advances in this field. Our 2024 collection of manuscripts showcases algorithmic advances that integrate data from distinct modalities and introduce innovative approaches to extract new, medically relevant information from existing data. These evolving technology and analytical methods promise to bring closer the goals of precision medicine to improve health and increase lifespan.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075218","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Zoish: A Novel Feature Selection Approach Leveraging Shapley Additive Values for Machine Learning Applications in Healthcare. Zoish:利用 Shapley 加法值的新特征选择方法,用于医疗保健领域的机器学习应用。
Hossein Javedani Sadaei, Salvatore Loguercio, Mahdi Shafiei Neyestanak, Ali Torkamani, Daria Prilutsky

In the intricate landscape of healthcare analytics, effective feature selection is a prerequisite for generating robust predictive models, especially given the common challenges of sample sizes and potential biases. Zoish uniquely addresses these issues by employing Shapley additive values-an idea rooted in cooperative game theory-to enable both transparent and automated feature selection. Unlike existing tools, Zoish is versatile, designed to seamlessly integrate with an array of machine learning libraries including scikit-learn, XGBoost, CatBoost, and imbalanced-learn.The distinct advantage of Zoish lies in its dual algorithmic approach for calculating Shapley values, allowing it to efficiently manage both large and small datasets. This adaptability renders it exceptionally suitable for a wide spectrum of healthcare-related tasks. The tool also places a strong emphasis on interpretability, providing comprehensive visualizations for analyzed features. Its customizable settings offer users fine-grained control over feature selection, thus optimizing for specific predictive objectives.This manuscript elucidates the mathematical framework underpinning Zoish and how it uniquely combines local and global feature selection into a single, streamlined process. To validate Zoish's efficiency and adaptability, we present case studies in breast cancer prediction and Montreal Cognitive Assessment (MoCA) prediction in Parkinson's disease, along with evaluations on 300 synthetic datasets. These applications underscore Zoish's unparalleled performance in diverse healthcare contexts and against its counterparts.

在错综复杂的医疗分析领域,有效的特征选择是生成稳健预测模型的先决条件,尤其是考虑到样本量和潜在偏差等常见挑战。Zoish 采用夏普利加法值(Shapley additive values)--一种植根于合作博弈论的理念--实现了透明和自动的特征选择,从而独特地解决了这些问题。与现有工具不同的是,Zoish 具有多功能性,可与一系列机器学习库无缝集成,包括 scikit-learn、XGBoost、CatBoost 和 imbalanced-learn。这种适应性使其非常适合广泛的医疗保健相关任务。该工具还非常注重可解释性,为分析特征提供全面的可视化效果。本手稿阐明了 Zoish 的数学框架,以及它如何将局部和全局特征选择独特地结合到一个单一、精简的流程中。为了验证 Zoish 的效率和适应性,我们介绍了乳腺癌预测和帕金森病蒙特利尔认知评估(MoCA)预测的案例研究,以及对 300 个合成数据集的评估。这些应用凸显了 Zoish 在不同医疗环境中与同行相比无与伦比的性能。
{"title":"Zoish: A Novel Feature Selection Approach Leveraging Shapley Additive Values for Machine Learning Applications in Healthcare.","authors":"Hossein Javedani Sadaei, Salvatore Loguercio, Mahdi Shafiei Neyestanak, Ali Torkamani, Daria Prilutsky","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>In the intricate landscape of healthcare analytics, effective feature selection is a prerequisite for generating robust predictive models, especially given the common challenges of sample sizes and potential biases. Zoish uniquely addresses these issues by employing Shapley additive values-an idea rooted in cooperative game theory-to enable both transparent and automated feature selection. Unlike existing tools, Zoish is versatile, designed to seamlessly integrate with an array of machine learning libraries including scikit-learn, XGBoost, CatBoost, and imbalanced-learn.The distinct advantage of Zoish lies in its dual algorithmic approach for calculating Shapley values, allowing it to efficiently manage both large and small datasets. This adaptability renders it exceptionally suitable for a wide spectrum of healthcare-related tasks. The tool also places a strong emphasis on interpretability, providing comprehensive visualizations for analyzed features. Its customizable settings offer users fine-grained control over feature selection, thus optimizing for specific predictive objectives.This manuscript elucidates the mathematical framework underpinning Zoish and how it uniquely combines local and global feature selection into a single, streamlined process. To validate Zoish's efficiency and adaptability, we present case studies in breast cancer prediction and Montreal Cognitive Assessment (MoCA) prediction in Parkinson's disease, along with evaluations on 300 synthetic datasets. These applications underscore Zoish's unparalleled performance in diverse healthcare contexts and against its counterparts.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10764073/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
FedBrain: Federated Training of Graph Neural Networks for Connectome-based Brain Imaging Analysis. FedBrain:基于连接体的脑成像分析的图神经网络联合训练。
Yi Yang, Han Xie, Hejie Cui, Carl Yang

Recent advancements in neuroimaging techniques have sparked a growing interest in understanding the complex interactions between anatomical regions of interest (ROIs), forming into brain networks that play a crucial role in various clinical tasks, such as neural pattern discovery and disorder diagnosis. In recent years, graph neural networks (GNNs) have emerged as powerful tools for analyzing network data. However, due to the complexity of data acquisition and regulatory restrictions, brain network studies remain limited in scale and are often confined to local institutions. These limitations greatly challenge GNN models to capture useful neural circuitry patterns and deliver robust downstream performance. As a distributed machine learning paradigm, federated learning (FL) provides a promising solution in addressing resource limitation and privacy concerns, by enabling collaborative learning across local institutions (i.e., clients) without data sharing. While the data heterogeneity issues have been extensively studied in recent FL literature, cross-institutional brain network analysis presents unique data heterogeneity challenges, that is, the inconsistent ROI parcellation systems and varying predictive neural circuitry patterns across local neuroimaging studies. To this end, we propose FedBrain, a GNN-based personalized FL framework that takes into account the unique properties of brain network data. Specifically, we present a federated atlas mapping mechanism to overcome the feature and structure heterogeneity of brain networks arising from different ROI atlas systems, and a clustering approach guided by clinical prior knowledge to address varying predictive neural circuitry patterns regarding different patient groups, neuroimaging modalities and clinical outcomes. Compared to existing FL strategies, our approach demonstrates superior and more consistent performance, showcasing its strong potential and generalizability in cross-institutional connectome-based brain imaging analysis. The implementation is available here.

神经成像技术的最新进展引发了人们对了解解剖学感兴趣区(ROIs)之间复杂相互作用的日益浓厚的兴趣,这些相互作用形成的大脑网络在神经模式发现和疾病诊断等各种临床任务中发挥着至关重要的作用。近年来,图神经网络(GNN)已成为分析网络数据的强大工具。然而,由于数据采集的复杂性和监管限制,脑网络研究的规模仍然有限,而且往往局限于本地机构。这些限制极大地挑战了 GNN 模型捕捉有用神经回路模式并提供稳健下游性能的能力。作为一种分布式机器学习范例,联合学习(FL)提供了一种很有前景的解决方案,它能在不共享数据的情况下,实现本地机构(即客户)之间的协作学习,从而解决资源限制和隐私问题。虽然数据异构问题已在最近的联合学习文献中得到了广泛研究,但跨机构脑网络分析面临着独特的数据异构挑战,即本地神经影像研究中不一致的 ROI 剖分系统和不同的预测神经回路模式。为此,我们提出了基于 GNN 的个性化 FL 框架 FedBrain,该框架考虑到了脑网络数据的独特属性。具体来说,我们提出了一种联合图集映射机制,以克服不同 ROI 图集系统产生的脑网络特征和结构异质性,并提出了一种以临床先验知识为指导的聚类方法,以解决不同患者群体、神经成像模式和临床结果的不同预测神经回路模式。与现有的 FL 策略相比,我们的方法表现出更优越、更稳定的性能,展示了其在跨机构基于连接体的脑成像分析中的强大潜力和通用性。具体实施请点击此处。
{"title":"FedBrain: Federated Training of Graph Neural Networks for Connectome-based Brain Imaging Analysis.","authors":"Yi Yang, Han Xie, Hejie Cui, Carl Yang","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Recent advancements in neuroimaging techniques have sparked a growing interest in understanding the complex interactions between anatomical regions of interest (ROIs), forming into brain networks that play a crucial role in various clinical tasks, such as neural pattern discovery and disorder diagnosis. In recent years, graph neural networks (GNNs) have emerged as powerful tools for analyzing network data. However, due to the complexity of data acquisition and regulatory restrictions, brain network studies remain limited in scale and are often confined to local institutions. These limitations greatly challenge GNN models to capture useful neural circuitry patterns and deliver robust downstream performance. As a distributed machine learning paradigm, federated learning (FL) provides a promising solution in addressing resource limitation and privacy concerns, by enabling collaborative learning across local institutions (i.e., clients) without data sharing. While the data heterogeneity issues have been extensively studied in recent FL literature, cross-institutional brain network analysis presents unique data heterogeneity challenges, that is, the inconsistent ROI parcellation systems and varying predictive neural circuitry patterns across local neuroimaging studies. To this end, we propose FedBrain, a GNN-based personalized FL framework that takes into account the unique properties of brain network data. Specifically, we present a federated atlas mapping mechanism to overcome the feature and structure heterogeneity of brain networks arising from different ROI atlas systems, and a clustering approach guided by clinical prior knowledge to address varying predictive neural circuitry patterns regarding different patient groups, neuroimaging modalities and clinical outcomes. Compared to existing FL strategies, our approach demonstrates superior and more consistent performance, showcasing its strong potential and generalizability in cross-institutional connectome-based brain imaging analysis. The implementation is available here.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075169","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Machine Learning Strategies for Improved Phenotype Prediction in Underrepresented Populations. 在代表性不足的人群中改进表型预测的机器学习策略。
David Bonet, May Levin, Daniel Mas Montserrat, Alexander G Ioannidis

Precision medicine models often perform better for populations of European ancestry due to the over-representation of this group in the genomic datasets and large-scale biobanks from which the models are constructed. As a result, prediction models may misrepresent or provide less accurate treatment recommendations for underrepresented populations, contributing to health disparities. This study introduces an adaptable machine learning toolkit that integrates multiple existing methodologies and novel techniques to enhance the prediction accuracy for underrepresented populations in genomic datasets. By leveraging machine learning techniques, including gradient boosting and automated methods, coupled with novel population-conditional re-sampling techniques, our method significantly improves the phenotypic prediction from single nucleotide polymorphism (SNP) data for diverse populations. We evaluate our approach using the UK Biobank, which is composed primarily of British individuals with European ancestry, and a minority representation of groups with Asian and African ancestry. Performance metrics demonstrate substantial improvements in phenotype prediction for underrepresented groups, achieving prediction accuracy comparable to that of the majority group. This approach represents a significant step towards improving prediction accuracy amidst current dataset diversity challenges. By integrating a tailored pipeline, our approach fosters more equitable validity and utility of statistical genetics methods, paving the way for more inclusive models and outcomes.

精准医学模型通常对欧洲血统的人群效果更好,这是因为在构建模型的基因组数据集和大规模生物库中,欧洲血统的人群所占比例过高。因此,预测模型可能会误导代表性不足的人群或为其提供不那么准确的治疗建议,从而造成健康差异。本研究介绍了一种可调整的机器学习工具包,该工具包整合了多种现有方法和新技术,以提高基因组数据集中代表性不足人群的预测准确性。通过利用梯度提升和自动化方法等机器学习技术,再加上新颖的人群条件再采样技术,我们的方法显著提高了单核苷酸多态性(SNP)数据对不同人群的表型预测。我们使用英国生物数据库对我们的方法进行了评估,该数据库主要由具有欧洲血统的英国人以及少数具有亚洲和非洲血统的群体组成。性能指标表明,对代表性不足群体的表型预测有了很大改进,预测准确率可与多数群体的预测准确率相媲美。在当前数据集多样性面临挑战的情况下,这种方法在提高预测准确性方面迈出了重要一步。通过整合量身定制的管道,我们的方法促进了统计遗传学方法更公平的有效性和实用性,为更具包容性的模型和结果铺平了道路。
{"title":"Machine Learning Strategies for Improved Phenotype Prediction in Underrepresented Populations.","authors":"David Bonet, May Levin, Daniel Mas Montserrat, Alexander G Ioannidis","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Precision medicine models often perform better for populations of European ancestry due to the over-representation of this group in the genomic datasets and large-scale biobanks from which the models are constructed. As a result, prediction models may misrepresent or provide less accurate treatment recommendations for underrepresented populations, contributing to health disparities. This study introduces an adaptable machine learning toolkit that integrates multiple existing methodologies and novel techniques to enhance the prediction accuracy for underrepresented populations in genomic datasets. By leveraging machine learning techniques, including gradient boosting and automated methods, coupled with novel population-conditional re-sampling techniques, our method significantly improves the phenotypic prediction from single nucleotide polymorphism (SNP) data for diverse populations. We evaluate our approach using the UK Biobank, which is composed primarily of British individuals with European ancestry, and a minority representation of groups with Asian and African ancestry. Performance metrics demonstrate substantial improvements in phenotype prediction for underrepresented groups, achieving prediction accuracy comparable to that of the majority group. This approach represents a significant step towards improving prediction accuracy amidst current dataset diversity challenges. By integrating a tailored pipeline, our approach fosters more equitable validity and utility of statistical genetics methods, paving the way for more inclusive models and outcomes.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10799683/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PEPSI: Polarity measurements from spatial proteomics imaging suggest immune cell engagement. PEPSI:空间蛋白质组学成像的极性测量表明免疫细胞参与其中。
Eric Wu, Zhenqin Wu, Aaron T Mayer, Alexandro E Trevino, James Zou

Subcellular protein localization is important for understanding functional states of cells, but measuring and quantifying this information can be difficult and typically requires high-resolution microscopy. In this work, we develop a metric to define surface protein polarity from immunofluorescence (IF) imaging data and use it to identify distinct immune cell states within tumor microenvironments. We apply this metric to characterize over two million cells across 600 patient samples and find that cells identified as having polar expression exhibit characteristics relating to tumor-immune cell engagement. Additionally, we show that incorporating these polarity-defined cell subtypes improves the performance of deep learning models trained to predict patient survival outcomes. This method provides a first look at using subcellular protein expression patterns to phenotype immune cell functional states with applications to precision medicine.

亚细胞蛋白质定位对了解细胞的功能状态非常重要,但测量和量化这些信息可能很困难,通常需要高分辨率的显微镜。在这项工作中,我们开发了一种度量方法,从免疫荧光(IF)成像数据中定义表面蛋白极性,并用它来识别肿瘤微环境中不同的免疫细胞状态。我们应用该指标对 600 份患者样本中的 200 多万个细胞进行了特征描述,发现被识别为具有极性表达的细胞表现出与肿瘤免疫细胞参与相关的特征。此外,我们还表明,结合这些极性定义的细胞亚型,可以提高为预测患者生存结果而训练的深度学习模型的性能。这种方法首次将亚细胞蛋白质表达模式用于表型免疫细胞功能状态,并应用于精准医疗。
{"title":"PEPSI: Polarity measurements from spatial proteomics imaging suggest immune cell engagement.","authors":"Eric Wu, Zhenqin Wu, Aaron T Mayer, Alexandro E Trevino, James Zou","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Subcellular protein localization is important for understanding functional states of cells, but measuring and quantifying this information can be difficult and typically requires high-resolution microscopy. In this work, we develop a metric to define surface protein polarity from immunofluorescence (IF) imaging data and use it to identify distinct immune cell states within tumor microenvironments. We apply this metric to characterize over two million cells across 600 patient samples and find that cells identified as having polar expression exhibit characteristics relating to tumor-immune cell engagement. Additionally, we show that incorporating these polarity-defined cell subtypes improves the performance of deep learning models trained to predict patient survival outcomes. This method provides a first look at using subcellular protein expression patterns to phenotype immune cell functional states with applications to precision medicine.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Spatial Omics Driven Crossmodal Pretraining Applied to Graph-based Deep Learning for Cancer Pathology Analysis. 将空间 Omics 驱动的跨模态预训练应用于癌症病理分析的基于图的深度学习。
Zarif L Azher, Michael Fatemi, Yunrui Lu, Gokul Srinivasan, Alos B Diallo, Brock C Christensen, Lucas A Salas, Fred W Kolling, Laurent Perreard, Scott M Palisoul, Louis J Vaickus, Joshua J Levy

Graph-based deep learning has shown great promise in cancer histopathology image analysis by contextualizing complex morphology and structure across whole slide images to make high quality downstream outcome predictions (ex: prognostication). These methods rely on informative representations (i.e., embeddings) of image patches comprising larger slides, which are used as node attributes in slide graphs. Spatial omics data, including spatial transcriptomics, is a novel paradigm offering a wealth of detailed information. Pairing this data with corresponding histological imaging localized at 50-micron resolution, may facilitate the development of algorithms which better appreciate the morphological and molecular underpinnings of carcinogenesis. Here, we explore the utility of leveraging spatial transcriptomics data with a contrastive crossmodal pretraining mechanism to generate deep learning models that can extract molecular and histological information for graph-based learning tasks. Performance on cancer staging, lymph node metastasis prediction, survival prediction, and tissue clustering analyses indicate that the proposed methods bring improvement to graph based deep learning models for histopathological slides compared to leveraging histological information from existing schemes, demonstrating the promise of mining spatial omics data to enhance deep learning for pathology workflows.

基于图的深度学习在癌症组织病理学图像分析中大有可为,它可以将整个切片图像中复杂的形态和结构上下文化,从而进行高质量的下游结果预测(例如:预后)。这些方法依赖于由较大切片组成的图像斑块的信息表征(即嵌入),这些斑块被用作切片图中的节点属性。空间 omics 数据(包括空间转录组学)是一种新型范例,可提供大量详细信息。将这些数据与以 50 微米分辨率定位的相应组织学成像配对,有助于开发能更好地了解癌变的形态学和分子基础的算法。在这里,我们探索了利用空间转录组学数据与对比性跨模态预训练机制生成深度学习模型的实用性,该模型可以为基于图的学习任务提取分子和组织学信息。在癌症分期、淋巴结转移预测、生存预测和组织聚类分析方面的表现表明,与利用现有方案中的组织学信息相比,所提出的方法为基于图的组织病理学切片深度学习模型带来了改进,证明了挖掘空间组学数据以增强病理学工作流深度学习的前景。
{"title":"Spatial Omics Driven Crossmodal Pretraining Applied to Graph-based Deep Learning for Cancer Pathology Analysis.","authors":"Zarif L Azher, Michael Fatemi, Yunrui Lu, Gokul Srinivasan, Alos B Diallo, Brock C Christensen, Lucas A Salas, Fred W Kolling, Laurent Perreard, Scott M Palisoul, Louis J Vaickus, Joshua J Levy","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Graph-based deep learning has shown great promise in cancer histopathology image analysis by contextualizing complex morphology and structure across whole slide images to make high quality downstream outcome predictions (ex: prognostication). These methods rely on informative representations (i.e., embeddings) of image patches comprising larger slides, which are used as node attributes in slide graphs. Spatial omics data, including spatial transcriptomics, is a novel paradigm offering a wealth of detailed information. Pairing this data with corresponding histological imaging localized at 50-micron resolution, may facilitate the development of algorithms which better appreciate the morphological and molecular underpinnings of carcinogenesis. Here, we explore the utility of leveraging spatial transcriptomics data with a contrastive crossmodal pretraining mechanism to generate deep learning models that can extract molecular and histological information for graph-based learning tasks. Performance on cancer staging, lymph node metastasis prediction, survival prediction, and tissue clustering analyses indicate that the proposed methods bring improvement to graph based deep learning models for histopathological slides compared to leveraging histological information from existing schemes, demonstrating the promise of mining spatial omics data to enhance deep learning for pathology workflows.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10783797/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evidence of recent and ongoing admixture in the U.S. and influences on health and disparities. 美国最近和正在发生的混血现象的证据以及对健康和差异的影响。
Hannah M Seagle, Jacklyn N Hellwege, Brian S Mautz, Chun Li, Yaomin Xu, Siwei Zhang, Dan M Roden, Tracy L McGregor, Digna R Velez Edwards, Todd L Edwards

Many researchers in genetics and social science incorporate information about race in their work. However, migrations (historical and forced) and social mobility have brought formerly separated populations of humans together, creating younger generations of individuals who have more complex and diverse ancestry and race profiles than older age groups. Here, we sought to better understand how temporal changes in genetic admixture influence levels of heterozygosity and impact health outcomes. We evaluated variation in genetic ancestry over 100 birth years in a cohort of 35,842 individuals with electronic health record (EHR) information in the Southeastern United States. Using the software STRUCTURE, we analyzed 2,678 ancestrally informative markers relative to three ancestral clusters (African, East Asian, and European) and observed rising levels of admixture for all clinically-defined race groups since 1990. Most race groups also exhibited increases in heterozygosity and long-range linkage disequilibrium over time, further supporting the finding of increasing admixture in young individuals in our cohort. These data are consistent with United States Census information from broader geographic areas and highlight the changing demography of the population. This increased diversity challenges classic approaches to studies of genotype-phenotype relationships which motivated us to explore the relationship between heterozygosity and disease diagnosis. Using a phenome-wide association study approach, we explored the relationship between admixture and disease risk and found that increased admixture resulted in protective associations with female reproductive disorders and increased risk for diseases with links to autoimmune dysfunction. These data suggest that tendencies in the United States population are increasing ancestral complexity over time. Further, these observations imply that, because both prevalence and severity of many diseases vary by race groups, complexity of ancestral origins influences health and disparities.

许多遗传学和社会科学研究人员在其工作中纳入了有关种族的信息。然而,(历史上的和被迫的)迁徙和社会流动将以前分离的人类群体聚集在一起,产生了年轻一代的个体,他们的祖先和种族特征比年龄较大的群体更为复杂和多样。在此,我们试图更好地了解基因混血的时间变化如何影响杂合度水平并对健康结果产生影响。我们评估了美国东南部 35,842 名有电子健康记录(EHR)信息的人在 100 个出生年中的遗传血统变化。利用 STRUCTURE 软件,我们分析了 2,678 个与三个祖先集群(非洲、东亚和欧洲)相关的祖先信息标记,观察到自 1990 年以来,所有临床定义的种族群体的混血水平都在上升。随着时间的推移,大多数种族群体的杂合度和长程连锁不平衡也在增加,这进一步支持了我们队列中年轻个体混血程度增加的发现。这些数据与美国更广泛地区的人口普查信息一致,凸显了人口结构的变化。多样性的增加对研究基因型与表型关系的传统方法提出了挑战,这促使我们探索杂合度与疾病诊断之间的关系。利用全表型关联研究方法,我们探讨了混血与疾病风险之间的关系,发现混血的增加导致女性生殖系统疾病的保护性关联,以及与自身免疫功能障碍有关的疾病风险增加。这些数据表明,随着时间的推移,美国人口的祖先复杂性有增加的趋势。此外,这些观察结果表明,由于许多疾病的发病率和严重程度因种族群体而异,祖先起源的复杂性影响着健康和差异。
{"title":"Evidence of recent and ongoing admixture in the U.S. and influences on health and disparities.","authors":"Hannah M Seagle, Jacklyn N Hellwege, Brian S Mautz, Chun Li, Yaomin Xu, Siwei Zhang, Dan M Roden, Tracy L McGregor, Digna R Velez Edwards, Todd L Edwards","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Many researchers in genetics and social science incorporate information about race in their work. However, migrations (historical and forced) and social mobility have brought formerly separated populations of humans together, creating younger generations of individuals who have more complex and diverse ancestry and race profiles than older age groups. Here, we sought to better understand how temporal changes in genetic admixture influence levels of heterozygosity and impact health outcomes. We evaluated variation in genetic ancestry over 100 birth years in a cohort of 35,842 individuals with electronic health record (EHR) information in the Southeastern United States. Using the software STRUCTURE, we analyzed 2,678 ancestrally informative markers relative to three ancestral clusters (African, East Asian, and European) and observed rising levels of admixture for all clinically-defined race groups since 1990. Most race groups also exhibited increases in heterozygosity and long-range linkage disequilibrium over time, further supporting the finding of increasing admixture in young individuals in our cohort. These data are consistent with United States Census information from broader geographic areas and highlight the changing demography of the population. This increased diversity challenges classic approaches to studies of genotype-phenotype relationships which motivated us to explore the relationship between heterozygosity and disease diagnosis. Using a phenome-wide association study approach, we explored the relationship between admixture and disease risk and found that increased admixture resulted in protective associations with female reproductive disorders and increased risk for diseases with links to autoimmune dysfunction. These data suggest that tendencies in the United States population are increasing ancestral complexity over time. Further, these observations imply that, because both prevalence and severity of many diseases vary by race groups, complexity of ancestral origins influences health and disparities.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075246","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Modeling Path Importance for Effective Alzheimer's Disease Drug Repurposing. 为有效的阿尔茨海默病药物再利用建立路径重要性模型
Shunian Xiang, Patrick J Lawrence, Bo Peng, ChienWei Chiang, Dokyoon Kim, Li Shen, Xia Ning

Recently, drug repurposing has emerged as an effective and resource-efficient paradigm for AD drug discovery. Among various methods for drug repurposing, network-based methods have shown promising results as they are capable of leveraging complex networks that integrate multiple interaction types, such as protein-protein interactions, to more effectively identify candidate drugs. However, existing approaches typically assume paths of the same length in the network have equal importance in identifying the therapeutic effect of drugs. Other domains have found that same length paths do not necessarily have the same importance. Thus, relying on this assumption may be deleterious to drug repurposing attempts. In this work, we propose MPI (Modeling Path Importance), a novel network-based method for AD drug repurposing. MPI is unique in that it prioritizes important paths via learned node embeddings, which can effectively capture a network's rich structural information. Thus, leveraging learned embeddings allows MPI to effectively differentiate the importance among paths. We evaluate MPI against a commonly used baseline method that identifies anti-AD drug candidates primarily based on the shortest paths between drugs and AD in the network. We observe that among the top-50 ranked drugs, MPI prioritizes 20.0% more drugs with anti-AD evidence compared to the baseline. Finally, Cox proportional-hazard models produced from insurance claims data aid us in identifying the use of etodolac, nicotine, and BBB-crossing ACE-INHs as having a reduced risk of AD, suggesting such drugs may be viable candidates for repurposing and should be explored further in future studies.

近来,药物再利用已成为一种有效且节省资源的AD药物发现范例。在各种药物再利用方法中,基于网络的方法显示出良好的效果,因为它们能够利用整合了多种相互作用类型(如蛋白质-蛋白质相互作用)的复杂网络,更有效地确定候选药物。然而,现有方法通常假定网络中相同长度的路径在确定药物治疗效果方面具有同等重要性。其他领域的研究发现,相同长度的路径并不一定具有相同的重要性。因此,依赖这一假设可能会不利于药物再利用的尝试。在这项工作中,我们提出了 MPI(路径重要性建模),这是一种基于网络的新型 AD 药物再利用方法。MPI 的独特之处在于,它通过学习的节点嵌入对重要路径进行优先排序,从而有效捕捉网络的丰富结构信息。因此,利用学习到的嵌入信息,MPI 可以有效区分不同路径的重要性。我们将 MPI 与一种常用的基线方法进行了对比评估,后者主要根据网络中药物与 AD 之间的最短路径来识别抗 AD 候选药物。我们发现,与基线方法相比,在排名前 50 位的药物中,MPI 优先选择的具有抗 AD 证据的药物多出 20.0%。最后,根据保险理赔数据建立的 Cox 比例危险模型帮助我们确定了使用依托度酸、尼古丁和跨越 BBB 的 ACE-INHs 可降低 AD 风险,这表明此类药物可能是再利用的可行候选药物,应在未来的研究中进一步探讨。
{"title":"Modeling Path Importance for Effective Alzheimer's Disease Drug Repurposing.","authors":"Shunian Xiang, Patrick J Lawrence, Bo Peng, ChienWei Chiang, Dokyoon Kim, Li Shen, Xia Ning","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Recently, drug repurposing has emerged as an effective and resource-efficient paradigm for AD drug discovery. Among various methods for drug repurposing, network-based methods have shown promising results as they are capable of leveraging complex networks that integrate multiple interaction types, such as protein-protein interactions, to more effectively identify candidate drugs. However, existing approaches typically assume paths of the same length in the network have equal importance in identifying the therapeutic effect of drugs. Other domains have found that same length paths do not necessarily have the same importance. Thus, relying on this assumption may be deleterious to drug repurposing attempts. In this work, we propose MPI (Modeling Path Importance), a novel network-based method for AD drug repurposing. MPI is unique in that it prioritizes important paths via learned node embeddings, which can effectively capture a network's rich structural information. Thus, leveraging learned embeddings allows MPI to effectively differentiate the importance among paths. We evaluate MPI against a commonly used baseline method that identifies anti-AD drug candidates primarily based on the shortest paths between drugs and AD in the network. We observe that among the top-50 ranked drugs, MPI prioritizes 20.0% more drugs with anti-AD evidence compared to the baseline. Finally, Cox proportional-hazard models produced from insurance claims data aid us in identifying the use of etodolac, nicotine, and BBB-crossing ACE-INHs as having a reduced risk of AD, suggesting such drugs may be viable candidates for repurposing and should be explored further in future studies.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11056095/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075191","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Imputation of race and ethnicity categories using genetic ancestry from real-world genomic testing data. 利用真实世界基因组测试数据中的遗传祖先推算种族和人种类别。
Brooke Rhead, Paige E Haffener, Yannick Pouliot, Francisco M De La Vega

The incompleteness of race and ethnicity information in real-world data (RWD) hampers its utility in promoting healthcare equity. This study introduces two methods-one heuristic and the other machine learning-based-to impute race and ethnicity from genetic ancestry using tumor profiling data. Analyzing de-identified data from over 100,000 cancer patients sequenced with the Tempus xT panel, we demonstrate that both methods outperform existing geolocation and surname-based methods, with the machine learning approach achieving high recall (range: 0.859-0.993) and precision (range: 0.932-0.981) across four mutually exclusive race and ethnicity categories. This work presents a novel pathway to enhance RWD utility in studying racial disparities in healthcare.

真实世界数据(RWD)中种族和民族信息的不完整性阻碍了其在促进医疗公平方面的作用。本研究介绍了两种方法--一种是启发式方法,另一种是基于机器学习的方法--利用肿瘤图谱数据从遗传祖先推算种族和人种。通过分析用 Tempus xT 面板测序的 10 万多名癌症患者的去标识化数据,我们证明这两种方法都优于现有的基于地理位置和姓氏的方法,其中机器学习方法在四个相互排斥的种族和民族类别中实现了高召回率(范围:0.859-0.993)和高精确度(范围:0.932-0.981)。这项工作提出了一种新的途径,以提高 RWD 在研究医疗保健中种族差异方面的效用。
{"title":"Imputation of race and ethnicity categories using genetic ancestry from real-world genomic testing data.","authors":"Brooke Rhead, Paige E Haffener, Yannick Pouliot, Francisco M De La Vega","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>The incompleteness of race and ethnicity information in real-world data (RWD) hampers its utility in promoting healthcare equity. This study introduces two methods-one heuristic and the other machine learning-based-to impute race and ethnicity from genetic ancestry using tumor profiling data. Analyzing de-identified data from over 100,000 cancer patients sequenced with the Tempus xT panel, we demonstrate that both methods outperform existing geolocation and surname-based methods, with the machine learning approach achieving high recall (range: 0.859-0.993) and precision (range: 0.932-0.981) across four mutually exclusive race and ethnicity categories. This work presents a novel pathway to enhance RWD utility in studying racial disparities in healthcare.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139075172","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing
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