首页 > 最新文献

Nature Machine Intelligence最新文献

英文 中文
Learning motif-based graphs for drug–drug interaction prediction via local–global self-attention 通过局部-全局自我关注,学习基于图案的药物相互作用预测图谱
IF 23.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-27 DOI: 10.1038/s42256-024-00888-6
Yi Zhong, Gaozheng Li, Ji Yang, Houbing Zheng, Yongqiang Yu, Jiheng Zhang, Heng Luo, Biao Wang, Zuquan Weng

Unexpected drug–drug interactions (DDIs) are important issues for both pharmaceutical research and clinical applications due to the high risk of causing severe adverse drug reactions or drug withdrawals. Many deep learning models have achieved high performance in DDI prediction, but model interpretability to reveal the underlying causes of DDIs has not been extensively explored. Here we propose MeTDDI—a deep learning framework with local–global self-attention and co-attention to learn motif-based graphs for DDI prediction. MeTDDI achieved competitive performance compared with state-of-the-art models. Regarding interpretability, we conducted extensive assessments on 73 drugs with 13,786 DDIs and MeTDDI can precisely explain the structural mechanisms for 5,602 DDIs involving 58 drugs. Besides, MeTDDI shows potential to explain complex DDI mechanisms and mitigate DDI risks. To summarize, MeTDDI provides a new perspective on exploring DDI mechanisms, which will benefit both drug discovery and polypharmacy for safer therapies for patients.

意外的药物相互作用(DDIs)是药物研究和临床应用中的重要问题,因为它极有可能导致严重的药物不良反应或停药。许多深度学习模型在 DDI 预测方面取得了很高的性能,但揭示 DDIs 潜在原因的模型可解释性尚未得到广泛探索。在此,我们提出了MeTDDI--一种具有局部-全局自关注和共关注的深度学习框架,用于学习基于主题图的DDI预测。与最先进的模型相比,MeTDDI 的性能极具竞争力。在可解释性方面,我们对73种药物的13786个DDIs进行了广泛评估,MeTDDI可以精确解释58种药物的5602个DDIs的结构机制。此外,MeTDDI 显示出解释复杂 DDI 机制和降低 DDI 风险的潜力。总之,MeTDDI 为探索 DDI 机制提供了一个新的视角,这将有利于药物发现和多药治疗,为患者提供更安全的疗法。
{"title":"Learning motif-based graphs for drug–drug interaction prediction via local–global self-attention","authors":"Yi Zhong, Gaozheng Li, Ji Yang, Houbing Zheng, Yongqiang Yu, Jiheng Zhang, Heng Luo, Biao Wang, Zuquan Weng","doi":"10.1038/s42256-024-00888-6","DOIUrl":"https://doi.org/10.1038/s42256-024-00888-6","url":null,"abstract":"<p>Unexpected drug–drug interactions (DDIs) are important issues for both pharmaceutical research and clinical applications due to the high risk of causing severe adverse drug reactions or drug withdrawals. Many deep learning models have achieved high performance in DDI prediction, but model interpretability to reveal the underlying causes of DDIs has not been extensively explored. Here we propose MeTDDI—a deep learning framework with local–global self-attention and co-attention to learn motif-based graphs for DDI prediction. MeTDDI achieved competitive performance compared with state-of-the-art models. Regarding interpretability, we conducted extensive assessments on 73 drugs with 13,786 DDIs and MeTDDI can precisely explain the structural mechanisms for 5,602 DDIs involving 58 drugs. Besides, MeTDDI shows potential to explain complex DDI mechanisms and mitigate DDI risks. To summarize, MeTDDI provides a new perspective on exploring DDI mechanisms, which will benefit both drug discovery and polypharmacy for safer therapies for patients.</p>","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":23.8,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142085189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A deep learning method that identifies cellular heterogeneity using nanoscale nuclear features 利用纳米级核特征识别细胞异质性的深度学习方法
IF 23.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-27 DOI: 10.1038/s42256-024-00883-x
Davide Carnevali, Limei Zhong, Esther González-Almela, Carlotta Viana, Mikhail Rotkevich, Aiping Wang, Daniel Franco-Barranco, Aitor Gonzalez-Marfil, Maria Victoria Neguembor, Alvaro Castells-Garcia, Ignacio Arganda-Carreras, Maria Pia Cosma

Cellular phenotypic heterogeneity is an important hallmark of many biological processes and understanding its origins remains a substantial challenge. This heterogeneity often reflects variations in the chromatin structure, influenced by factors such as viral infections and cancer, which dramatically reshape the cellular landscape. To address the challenge of identifying distinct cell states, we developed artificial intelligence of the nucleus (AINU), a deep learning method that can identify specific nuclear signatures at the nanoscale resolution. AINU can distinguish different cell states based on the spatial arrangement of core histone H3, RNA polymerase II or DNA from super-resolution microscopy images. With only a small number of images as the training data, AINU correctly identifies human somatic cells, human-induced pluripotent stem cells, very early stage infected cells transduced with DNA herpes simplex virus type 1 and even cancer cells after appropriate retraining. Finally, using AI interpretability methods, we find that the RNA polymerase II localizations in the nucleoli aid in distinguishing human-induced pluripotent stem cells from their somatic cells. Overall, AINU coupled with super-resolution microscopy of nuclear structures provides a robust tool for the precise detection of cellular heterogeneity, with considerable potential for advancing diagnostics and therapies in regenerative medicine, virology and cancer biology.

细胞表型异质性是许多生物过程的重要标志,而了解其起源仍是一项巨大的挑战。这种异质性通常反映了染色质结构的变化,受病毒感染和癌症等因素的影响,这些因素极大地重塑了细胞景观。为了应对识别不同细胞状态的挑战,我们开发了细胞核人工智能(AINU),这是一种深度学习方法,可以在纳米级分辨率上识别特定的核特征。AINU 可以根据超分辨率显微镜图像中核心组蛋白 H3、RNA 聚合酶 II 或 DNA 的空间排列来区分不同的细胞状态。AINU 仅用少量图像作为训练数据,就能正确识别人类体细胞、人类诱导多能干细胞、DNA 1 型单纯疱疹病毒转导的早期感染细胞,甚至在经过适当的再训练后还能识别癌细胞。最后,利用人工智能可解释性方法,我们发现核小体中 RNA 聚合酶 II 的定位有助于区分人类诱导多能干细胞和体细胞。总之,AINU 与核结构超分辨率显微镜相结合,为精确检测细胞异质性提供了一个强大的工具,在再生医学、病毒学和癌症生物学的诊断和治疗方面具有相当大的潜力。
{"title":"A deep learning method that identifies cellular heterogeneity using nanoscale nuclear features","authors":"Davide Carnevali, Limei Zhong, Esther González-Almela, Carlotta Viana, Mikhail Rotkevich, Aiping Wang, Daniel Franco-Barranco, Aitor Gonzalez-Marfil, Maria Victoria Neguembor, Alvaro Castells-Garcia, Ignacio Arganda-Carreras, Maria Pia Cosma","doi":"10.1038/s42256-024-00883-x","DOIUrl":"https://doi.org/10.1038/s42256-024-00883-x","url":null,"abstract":"<p>Cellular phenotypic heterogeneity is an important hallmark of many biological processes and understanding its origins remains a substantial challenge. This heterogeneity often reflects variations in the chromatin structure, influenced by factors such as viral infections and cancer, which dramatically reshape the cellular landscape. To address the challenge of identifying distinct cell states, we developed artificial intelligence of the nucleus (AINU), a deep learning method that can identify specific nuclear signatures at the nanoscale resolution. AINU can distinguish different cell states based on the spatial arrangement of core histone H3, RNA polymerase II or DNA from super-resolution microscopy images. With only a small number of images as the training data, AINU correctly identifies human somatic cells, human-induced pluripotent stem cells, very early stage infected cells transduced with DNA herpes simplex virus type 1 and even cancer cells after appropriate retraining. Finally, using AI interpretability methods, we find that the RNA polymerase II localizations in the nucleoli aid in distinguishing human-induced pluripotent stem cells from their somatic cells. Overall, AINU coupled with super-resolution microscopy of nuclear structures provides a robust tool for the precise detection of cellular heterogeneity, with considerable potential for advancing diagnostics and therapies in regenerative medicine, virology and cancer biology.</p>","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":23.8,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142085188","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Factuality challenges in the era of large language models and opportunities for fact-checking 大型语言模型时代的事实挑战与事实核查机遇
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-22 DOI: 10.1038/s42256-024-00881-z
Isabelle Augenstein, Timothy Baldwin, Meeyoung Cha, Tanmoy Chakraborty, Giovanni Luca Ciampaglia, David Corney, Renee DiResta, Emilio Ferrara, Scott Hale, Alon Halevy, Eduard Hovy, Heng Ji, Filippo Menczer, Ruben Miguez, Preslav Nakov, Dietram Scheufele, Shivam Sharma, Giovanni Zagni
The emergence of tools based on large language models (LLMs), such as OpenAI’s ChatGPT and Google’s Gemini, has garnered immense public attention owing to their advanced natural language generation capabilities. These remarkably natural-sounding tools have the potential to be highly useful for various tasks. However, they also tend to produce false, erroneous or misleading content—commonly referred to as hallucinations. Moreover, LLMs can be misused to generate convincing, yet false, content and profiles on a large scale, posing a substantial societal challenge by potentially deceiving users and spreading inaccurate information. This makes fact-checking increasingly important. Despite their issues with factual accuracy, LLMs have shown proficiency in various subtasks that support fact-checking, which is essential to ensure factually accurate responses. In light of these concerns, we explore issues related to factuality in LLMs and their impact on fact-checking. We identify key challenges, imminent threats and possible solutions to these factuality issues. We also thoroughly examine these challenges, existing solutions and potential prospects for fact-checking. By analysing the factuality constraints within LLMs and their impact on fact-checking, we aim to contribute to a path towards maintaining accuracy at a time of confluence of generative artificial intelligence and misinformation. Large language models (LLMs) present challenges, including a tendency to produce false or misleading content and the potential to create misinformation or disinformation. Augenstein and colleagues explore issues related to factuality in LLMs and their impact on fact-checking.
基于大型语言模型(LLM)的工具,如 OpenAI 的 ChatGPT 和谷歌的 Gemini,因其先进的自然语言生成能力而备受公众关注。这些听起来非常自然的工具有可能在各种任务中发挥巨大作用。然而,它们也容易产生虚假、错误或误导性的内容--通常被称为幻觉。此外,LLM 还可能被滥用,大规模生成令人信服的虚假内容和简介,从而可能欺骗用户并传播不准确的信息,对社会构成巨大挑战。因此,事实核查变得越来越重要。尽管法律硕士在事实准确性方面存在问题,但他们在各种支持事实检查的子任务中表现出了熟练的能力,这对于确保回复的事实准确性至关重要。鉴于这些问题,我们探讨了与法律硕士的事实准确性有关的问题及其对事实核查的影响。我们确定了这些事实真实性问题所面临的主要挑战、迫在眉睫的威胁和可能的解决方案。我们还深入研究了这些挑战、现有解决方案以及事实核查的潜在前景。通过分析法律文献中的事实性限制及其对事实核查的影响,我们旨在为在生成式人工智能和错误信息交织的时代保持准确性的道路做出贡献。
{"title":"Factuality challenges in the era of large language models and opportunities for fact-checking","authors":"Isabelle Augenstein,&nbsp;Timothy Baldwin,&nbsp;Meeyoung Cha,&nbsp;Tanmoy Chakraborty,&nbsp;Giovanni Luca Ciampaglia,&nbsp;David Corney,&nbsp;Renee DiResta,&nbsp;Emilio Ferrara,&nbsp;Scott Hale,&nbsp;Alon Halevy,&nbsp;Eduard Hovy,&nbsp;Heng Ji,&nbsp;Filippo Menczer,&nbsp;Ruben Miguez,&nbsp;Preslav Nakov,&nbsp;Dietram Scheufele,&nbsp;Shivam Sharma,&nbsp;Giovanni Zagni","doi":"10.1038/s42256-024-00881-z","DOIUrl":"10.1038/s42256-024-00881-z","url":null,"abstract":"The emergence of tools based on large language models (LLMs), such as OpenAI’s ChatGPT and Google’s Gemini, has garnered immense public attention owing to their advanced natural language generation capabilities. These remarkably natural-sounding tools have the potential to be highly useful for various tasks. However, they also tend to produce false, erroneous or misleading content—commonly referred to as hallucinations. Moreover, LLMs can be misused to generate convincing, yet false, content and profiles on a large scale, posing a substantial societal challenge by potentially deceiving users and spreading inaccurate information. This makes fact-checking increasingly important. Despite their issues with factual accuracy, LLMs have shown proficiency in various subtasks that support fact-checking, which is essential to ensure factually accurate responses. In light of these concerns, we explore issues related to factuality in LLMs and their impact on fact-checking. We identify key challenges, imminent threats and possible solutions to these factuality issues. We also thoroughly examine these challenges, existing solutions and potential prospects for fact-checking. By analysing the factuality constraints within LLMs and their impact on fact-checking, we aim to contribute to a path towards maintaining accuracy at a time of confluence of generative artificial intelligence and misinformation. Large language models (LLMs) present challenges, including a tendency to produce false or misleading content and the potential to create misinformation or disinformation. Augenstein and colleagues explore issues related to factuality in LLMs and their impact on fact-checking.","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142022075","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A bioactivity foundation model using pairwise meta-learning 使用成对元学习的生物活性基础模型
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-14 DOI: 10.1038/s42256-024-00876-w
Bin Feng, Zequn Liu, Nanlan Huang, Zhiping Xiao, Haomiao Zhang, Srbuhi Mirzoyan, Hanwen Xu, Jiaran Hao, Yinghui Xu, Ming Zhang, Sheng Wang
The bioactivity of compounds plays an important role in drug development and discovery. Existing machine learning approaches have poor generalizability in bioactivity prediction due to the small number of compounds in each assay and incompatible measurements among assays. In this paper, we propose ActFound, a bioactivity foundation model trained on 1.6 million experimentally measured bioactivities and 35,644 assays from ChEMBL. The key idea of ActFound is to use pairwise learning to learn the relative bioactivity differences between two compounds within the same assay to circumvent the incompatibility among assays. ActFound further exploits meta-learning to jointly optimize the model from all assays. On six real-world bioactivity datasets, ActFound demonstrates accurate in-domain prediction and strong generalization across assay types and molecular scaffolds. We also demonstrate that ActFound can be used as an accurate alternative to the leading physics-based computational tool FEP+(OPLS4) by achieving comparable performance when using only a few data points for fine-tuning. Our promising results indicate that ActFound could be an effective bioactivity foundation model for compound bioactivity prediction, paving the way for machine-learning-based drug development and discovery. Traditional machine learning methods for drug development struggle with bioactivity prediction due to the limited number of compounds in each assay and assay incompatibilities. Feng et al. developed ActFound, a bioactivity foundation model trained by pairwise learning and meta-learning, that improves the accuracy and generalization of bioactivity prediction.
化合物的生物活性在药物开发和发现中发挥着重要作用。现有的机器学习方法在生物活性预测方面的普适性较差,原因是每种检测方法中的化合物数量较少,且检测方法之间的测量结果不兼容。在本文中,我们提出了 ActFound,这是一种生物活性基础模型,它是在 160 万个实验测量的生物活性和来自 ChEMBL 的 35,644 种检测方法的基础上训练而成的。ActFound 的主要理念是利用成对学习来学习同一检测中两种化合物之间的相对生物活性差异,以规避检测之间的不兼容性。ActFound 还进一步利用元学习(meta-learning)来联合优化来自所有测定的模型。在六个真实世界的生物活性数据集上,ActFound 展示了准确的域内预测,以及跨测定类型和分子支架的强大泛化能力。我们还证明了 ActFound 可以作为领先的基于物理的计算工具 FEP+(OPLS4)的精确替代品,只需使用几个数据点进行微调,就能获得相当的性能。我们充满希望的结果表明,ActFound 可以成为化合物生物活性预测的有效生物活性基础模型,为基于机器学习的药物开发和发现铺平道路。
{"title":"A bioactivity foundation model using pairwise meta-learning","authors":"Bin Feng,&nbsp;Zequn Liu,&nbsp;Nanlan Huang,&nbsp;Zhiping Xiao,&nbsp;Haomiao Zhang,&nbsp;Srbuhi Mirzoyan,&nbsp;Hanwen Xu,&nbsp;Jiaran Hao,&nbsp;Yinghui Xu,&nbsp;Ming Zhang,&nbsp;Sheng Wang","doi":"10.1038/s42256-024-00876-w","DOIUrl":"10.1038/s42256-024-00876-w","url":null,"abstract":"The bioactivity of compounds plays an important role in drug development and discovery. Existing machine learning approaches have poor generalizability in bioactivity prediction due to the small number of compounds in each assay and incompatible measurements among assays. In this paper, we propose ActFound, a bioactivity foundation model trained on 1.6 million experimentally measured bioactivities and 35,644 assays from ChEMBL. The key idea of ActFound is to use pairwise learning to learn the relative bioactivity differences between two compounds within the same assay to circumvent the incompatibility among assays. ActFound further exploits meta-learning to jointly optimize the model from all assays. On six real-world bioactivity datasets, ActFound demonstrates accurate in-domain prediction and strong generalization across assay types and molecular scaffolds. We also demonstrate that ActFound can be used as an accurate alternative to the leading physics-based computational tool FEP+(OPLS4) by achieving comparable performance when using only a few data points for fine-tuning. Our promising results indicate that ActFound could be an effective bioactivity foundation model for compound bioactivity prediction, paving the way for machine-learning-based drug development and discovery. Traditional machine learning methods for drug development struggle with bioactivity prediction due to the limited number of compounds in each assay and assay incompatibilities. Feng et al. developed ActFound, a bioactivity foundation model trained by pairwise learning and meta-learning, that improves the accuracy and generalization of bioactivity prediction.","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141980824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
On responsible machine learning datasets emphasizing fairness, privacy and regulatory norms with examples in biometrics and healthcare 以生物识别和医疗保健领域为例,介绍强调公平、隐私和监管规范的负责任机器学习数据集
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-12 DOI: 10.1038/s42256-024-00874-y
Surbhi Mittal, Kartik Thakral, Richa Singh, Mayank Vatsa, Tamar Glaser, Cristian Canton Ferrer, Tal Hassner
Artificial Intelligence (AI) has seamlessly integrated into numerous scientific domains, catalysing unparalleled enhancements across a broad spectrum of tasks; however, its integrity and trustworthiness have emerged as notable concerns. The scientific community has focused on the development of trustworthy AI algorithms; however, machine learning and deep learning algorithms, popular in the AI community today, intrinsically rely on the quality of their training data. These algorithms are designed to detect patterns within the data, thereby learning the intended behavioural objectives. Any inadequacy in the data has the potential to translate directly into algorithms. In this study we discuss the importance of responsible machine learning datasets through the lens of fairness, privacy and regulatory compliance, and present a large audit of computer vision datasets. Despite the ubiquity of fairness and privacy challenges across diverse data domains, current regulatory frameworks primarily address human-centric data concerns. We therefore focus our discussion on biometric and healthcare datasets, although the principles we outline are broadly applicable across various domains. The audit is conducted through evaluation of the proposed responsible rubric. After surveying over 100 datasets, our detailed analysis of 60 distinct datasets highlights a universal susceptibility to fairness, privacy and regulatory compliance issues. This finding emphasizes the urgent need for revising dataset creation methodologies within the scientific community, especially in light of global advancements in data protection legislation. We assert that our study is critically relevant in the contemporary AI context, offering insights and recommendations that are both timely and essential for the ongoing evolution of AI technologies. There are pervasive concerns related to fairness, privacy and regulatory compliance in machine learning applications in healthcare, necessitating a reevaluation of dataset creation practices. Mittal et al. examine various computer vision datasets, providing insights to foster responsible AI development.
人工智能(AI)已无缝融入众多科学领域,在广泛的任务中催生了无与伦比的提升;然而,其完整性和可信度已成为值得关注的问题。科学界一直专注于开发值得信赖的人工智能算法;然而,当今人工智能界流行的机器学习和深度学习算法本质上依赖于其训练数据的质量。这些算法旨在检测数据中的模式,从而学习预期的行为目标。数据中的任何不足都有可能直接转化为算法。在本研究中,我们从公平、隐私和法规遵从的角度讨论了负责任的机器学习数据集的重要性,并对计算机视觉数据集进行了大规模审计。尽管公平性和隐私性挑战普遍存在于不同的数据领域,但当前的监管框架主要解决的是以人为中心的数据问题。因此,我们将讨论重点放在生物识别和医疗保健数据集上,尽管我们概述的原则广泛适用于各个领域。审计是通过评估建议的责任标准进行的。在对 100 多个数据集进行调查后,我们对 60 个不同的数据集进行了详细分析,结果表明,这些数据集普遍存在公平性、隐私性和法规遵从性问题。这一发现强调了在科学界修改数据集创建方法的迫切需要,尤其是在全球数据保护立法不断进步的情况下。我们断言,我们的研究与当代人工智能背景密切相关,为人工智能技术的不断发展提供了及时而重要的见解和建议。
{"title":"On responsible machine learning datasets emphasizing fairness, privacy and regulatory norms with examples in biometrics and healthcare","authors":"Surbhi Mittal,&nbsp;Kartik Thakral,&nbsp;Richa Singh,&nbsp;Mayank Vatsa,&nbsp;Tamar Glaser,&nbsp;Cristian Canton Ferrer,&nbsp;Tal Hassner","doi":"10.1038/s42256-024-00874-y","DOIUrl":"10.1038/s42256-024-00874-y","url":null,"abstract":"Artificial Intelligence (AI) has seamlessly integrated into numerous scientific domains, catalysing unparalleled enhancements across a broad spectrum of tasks; however, its integrity and trustworthiness have emerged as notable concerns. The scientific community has focused on the development of trustworthy AI algorithms; however, machine learning and deep learning algorithms, popular in the AI community today, intrinsically rely on the quality of their training data. These algorithms are designed to detect patterns within the data, thereby learning the intended behavioural objectives. Any inadequacy in the data has the potential to translate directly into algorithms. In this study we discuss the importance of responsible machine learning datasets through the lens of fairness, privacy and regulatory compliance, and present a large audit of computer vision datasets. Despite the ubiquity of fairness and privacy challenges across diverse data domains, current regulatory frameworks primarily address human-centric data concerns. We therefore focus our discussion on biometric and healthcare datasets, although the principles we outline are broadly applicable across various domains. The audit is conducted through evaluation of the proposed responsible rubric. After surveying over 100 datasets, our detailed analysis of 60 distinct datasets highlights a universal susceptibility to fairness, privacy and regulatory compliance issues. This finding emphasizes the urgent need for revising dataset creation methodologies within the scientific community, especially in light of global advancements in data protection legislation. We assert that our study is critically relevant in the contemporary AI context, offering insights and recommendations that are both timely and essential for the ongoing evolution of AI technologies. There are pervasive concerns related to fairness, privacy and regulatory compliance in machine learning applications in healthcare, necessitating a reevaluation of dataset creation practices. Mittal et al. examine various computer vision datasets, providing insights to foster responsible AI development.","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s42256-024-00874-y.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141974035","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Data-driven discovery of movement-linked heterogeneity in neurodegenerative diseases 数据驱动发现神经退行性疾病中与运动相关的异质性
IF 23.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-09 DOI: 10.1038/s42256-024-00882-y
Mark Endo, Favour Nerrise, Qingyu Zhao, Edith V. Sullivan, Li Fei-Fei, Victor W. Henderson, Kilian M. Pohl, Kathleen L. Poston, Ehsan Adeli

Neurodegenerative diseases manifest different motor and cognitive signs and symptoms that are highly heterogeneous. Parsing these heterogeneities may lead to an improved understanding of underlying disease mechanisms; however, current methods are dependent on clinical assessments and an arbitrary choice of behavioural tests. Here we present a data-driven subtyping approach using video-captured human motion and brain functional connectivity from resting-state functional magnetic resonance imaging. We applied our framework to a cohort of individuals at different stages of Parkinson’s disease. The process mapped the data to low-dimensional measures by projecting them onto a canonical correlation space that identified three Parkinson’s disease subtypes: subtype I was characterized by motor difficulties and poor visuospatial abilities; subtype II exhibited difficulties in non-motor components of activities of daily living and motor complications (dyskinesias and motor fluctuations) and subtype III was characterized by predominant tremor symptoms. We conducted a convergent validity analysis by comparing our approach to existing and widely used approaches. The compared approaches yielded subtypes that were adequately well-clustered in the motion-brain representation space we created to delineate subtypes. Our data-driven approach, contrary to other forms of subtyping, derived biomarkers predictive of motion impairment and subtype memberships that were captured objectively by digital videos.

神经退行性疾病表现出不同的运动和认知体征和症状,具有高度异质性。解析这些异质性可能有助于更好地了解潜在的疾病机制;然而,目前的方法依赖于临床评估和任意选择的行为测试。在这里,我们提出了一种数据驱动的亚型分析方法,该方法使用视频捕捉的人体运动和静息状态功能磁共振成像的大脑功能连接。我们将这一框架应用于处于帕金森病不同阶段的人群。这一过程通过将数据投射到一个典型相关空间,将数据映射到低维测量值,从而确定了三种帕金森病亚型:亚型 I 的特征是运动困难和视觉空间能力差;亚型 II 表现出日常生活活动中的非运动部分困难和运动并发症(运动障碍和运动波动);亚型 III 的特征是震颤症状占主导地位。通过将我们的方法与现有的、广泛使用的方法进行比较,我们进行了收敛有效性分析。比较后得出的亚型在我们为划分亚型而创建的运动-大脑表征空间中充分聚类。与其他形式的亚型划分方法不同,我们的数据驱动方法通过数字视频客观地捕捉到了可预测运动障碍和亚型成员的生物标志物。
{"title":"Data-driven discovery of movement-linked heterogeneity in neurodegenerative diseases","authors":"Mark Endo, Favour Nerrise, Qingyu Zhao, Edith V. Sullivan, Li Fei-Fei, Victor W. Henderson, Kilian M. Pohl, Kathleen L. Poston, Ehsan Adeli","doi":"10.1038/s42256-024-00882-y","DOIUrl":"https://doi.org/10.1038/s42256-024-00882-y","url":null,"abstract":"<p>Neurodegenerative diseases manifest different motor and cognitive signs and symptoms that are highly heterogeneous. Parsing these heterogeneities may lead to an improved understanding of underlying disease mechanisms; however, current methods are dependent on clinical assessments and an arbitrary choice of behavioural tests. Here we present a data-driven subtyping approach using video-captured human motion and brain functional connectivity from resting-state functional magnetic resonance imaging. We applied our framework to a cohort of individuals at different stages of Parkinson’s disease. The process mapped the data to low-dimensional measures by projecting them onto a canonical correlation space that identified three Parkinson’s disease subtypes: subtype I was characterized by motor difficulties and poor visuospatial abilities; subtype II exhibited difficulties in non-motor components of activities of daily living and motor complications (dyskinesias and motor fluctuations) and subtype III was characterized by predominant tremor symptoms. We conducted a convergent validity analysis by comparing our approach to existing and widely used approaches. The compared approaches yielded subtypes that were adequately well-clustered in the motion-brain representation space we created to delineate subtypes. Our data-driven approach, contrary to other forms of subtyping, derived biomarkers predictive of motion impairment and subtype memberships that were captured objectively by digital videos.</p>","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":23.8,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141908969","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cognitive maps from predictive vision 来自预测视觉的认知地图
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-08 DOI: 10.1038/s42256-024-00885-9
Margaret C. von Ebers, Xue-Xin Wei
Constructing spatial maps from sensory inputs is challenging in both neuroscience and artificial intelligence. A recent study demonstrates that a self-attention neural network using predictive coding can generate an environmental map in its latent space as an agent that navigates the environment.
从感觉输入构建空间地图在神经科学和人工智能领域都具有挑战性。最近的一项研究表明,使用预测编码的自我注意神经网络可以在其潜在空间中生成环境地图,成为一个在环境中导航的代理。
{"title":"Cognitive maps from predictive vision","authors":"Margaret C. von Ebers,&nbsp;Xue-Xin Wei","doi":"10.1038/s42256-024-00885-9","DOIUrl":"10.1038/s42256-024-00885-9","url":null,"abstract":"Constructing spatial maps from sensory inputs is challenging in both neuroscience and artificial intelligence. A recent study demonstrates that a self-attention neural network using predictive coding can generate an environmental map in its latent space as an agent that navigates the environment.","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141904419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrated structure prediction of protein–protein docking with experimental restraints using ColabDock 利用 ColabDock 进行带有实验约束的蛋白质-蛋白质对接的综合结构预测
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-05 DOI: 10.1038/s42256-024-00873-z
Shihao Feng, Zhenyu Chen, Chengwei Zhang, Yuhao Xie, Sergey Ovchinnikov, Yi Qin Gao, Sirui Liu
Protein complex structure prediction plays important roles in various applications, such as drug discovery and antibody design. However, due to limited prediction accuracy, there are frequent inconsistencies between the predictions and the experiments. Here we present ColabDock, a general framework adapting deep learning structure prediction models to integrate experimental restraints of different forms and sources without further large-scale retraining or fine tuning. With a generation–prediction architecture and trained ranking model, ColabDock outperforms HADDOCK and ClusPro using AlphaFold2 as the structure prediction model, not only in complex structure predictions with simulated residue and surface restraints but also in those assisted by nuclear magnetic resonance chemical shift perturbation as well as covalent labelling. It also assists antibody–antigen interface prediction with emulated interface scan restraints, which could be obtained by experiments such as deep mutational scanning. As a unified framework, we hope that ColabDock can help to bridge the gap between experimental and computational protein science. Despite rapid developments in predicting the complex structures of proteins, there are still inconsistencies between predictions and experiments. Feng et al. developed ColabDock, a general framework for deep learning models that integrates various experimental restraints and improves complex interface prediction, including antibody–antigen interactions.
蛋白质复合物结构预测在药物发现和抗体设计等各种应用中发挥着重要作用。然而,由于预测精度有限,预测与实验之间经常出现不一致。在此,我们提出了一个通用框架 ColabDock,该框架调整了深度学习结构预测模型,以整合不同形式和来源的实验约束,而无需进一步的大规模再训练或微调。利用生成-预测架构和训练有素的排序模型,ColabDock不仅在模拟残基和表面约束的复杂结构预测方面优于HADDOCK和使用AlphaFold2作为结构预测模型的ClusPro,而且在核磁共振化学位移扰动和共价标记辅助的复杂结构预测方面也优于HADDOCK和ClusPro。它还能利用仿真界面扫描约束条件辅助抗体-抗原界面预测,这些约束条件可通过深度突变扫描等实验获得。作为一个统一的框架,我们希望 ColabDock 能够帮助弥合实验和计算蛋白质科学之间的差距。
{"title":"Integrated structure prediction of protein–protein docking with experimental restraints using ColabDock","authors":"Shihao Feng,&nbsp;Zhenyu Chen,&nbsp;Chengwei Zhang,&nbsp;Yuhao Xie,&nbsp;Sergey Ovchinnikov,&nbsp;Yi Qin Gao,&nbsp;Sirui Liu","doi":"10.1038/s42256-024-00873-z","DOIUrl":"10.1038/s42256-024-00873-z","url":null,"abstract":"Protein complex structure prediction plays important roles in various applications, such as drug discovery and antibody design. However, due to limited prediction accuracy, there are frequent inconsistencies between the predictions and the experiments. Here we present ColabDock, a general framework adapting deep learning structure prediction models to integrate experimental restraints of different forms and sources without further large-scale retraining or fine tuning. With a generation–prediction architecture and trained ranking model, ColabDock outperforms HADDOCK and ClusPro using AlphaFold2 as the structure prediction model, not only in complex structure predictions with simulated residue and surface restraints but also in those assisted by nuclear magnetic resonance chemical shift perturbation as well as covalent labelling. It also assists antibody–antigen interface prediction with emulated interface scan restraints, which could be obtained by experiments such as deep mutational scanning. As a unified framework, we hope that ColabDock can help to bridge the gap between experimental and computational protein science. Despite rapid developments in predicting the complex structures of proteins, there are still inconsistencies between predictions and experiments. Feng et al. developed ColabDock, a general framework for deep learning models that integrates various experimental restraints and improves complex interface prediction, including antibody–antigen interactions.","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141895210","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Author Correction: A 5′ UTR language model for decoding untranslated regions of mRNA and function predictions 作者更正:用于解码 mRNA 非翻译区和功能预测的 5′ UTR 语言模型
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-08-02 DOI: 10.1038/s42256-024-00890-y
Yanyi Chu, Dan Yu, Yupeng Li, Kaixuan Huang, Yue Shen, Le Cong, Jason Zhang, Mengdi Wang
{"title":"Author Correction: A 5′ UTR language model for decoding untranslated regions of mRNA and function predictions","authors":"Yanyi Chu,&nbsp;Dan Yu,&nbsp;Yupeng Li,&nbsp;Kaixuan Huang,&nbsp;Yue Shen,&nbsp;Le Cong,&nbsp;Jason Zhang,&nbsp;Mengdi Wang","doi":"10.1038/s42256-024-00890-y","DOIUrl":"10.1038/s42256-024-00890-y","url":null,"abstract":"","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s42256-024-00890-y.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142091165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Deep learning prediction of glycopeptide tandem mass spectra powers glycoproteomics 糖肽串联质谱的深度学习预测为糖蛋白组学提供动力
IF 18.8 1区 计算机科学 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-30 DOI: 10.1038/s42256-024-00875-x
Yu Zong, Yuxin Wang, Xipeng Qiu, Xuanjing Huang, Liang Qiao
Protein glycosylation, a post-translational modification of proteins by glycans, plays an important role in numerous physiological and pathological cellular functions. Glycoproteomics, the study of protein glycosylation on a proteome-wide scale, utilizes liquid chromatography coupled with tandem mass spectrometry (MS/MS) to get combinational information on glycosylation site, glycosylation level and glycan structure. However, current database searching methods for glycoproteomics often struggle with glycan structure determination due to the limited occurrence of structure-determining ions. Although spectral searching methods can leverage fragment intensity to facilitate the structure identification of glycopeptides, their application is hindered by difficulties in spectral library construction. In this work, we present DeepGP, a hybrid deep learning framework based on transformer and graph neural networks, for the prediction of MS/MS spectra and retention time of glycopeptides. Two graph neural network modules are employed to capture the branched glycan structure and predict glycan ion intensity, respectively. Additionally, a pretraining strategy is implemented to alleviate the insufficiency of glycoproteomics data. Testing on multiple biological datasets, DeepGP accurately predicts MS/MS spectra and retention time of glycopeptides, closely aligning with the experimental results. Comprehensive benchmarking of DeepGP on synthetic and biological datasets validates its effectiveness in distinguishing similar glycans. Based on various decoy methods, DeepGP in combination with database searching can increase glycopeptide detection sensitivity. We anticipate that DeepGP can inspire extensive future work in glycoproteomics. Glycosylation, a prevalent type of post-translational modification of proteins by glycan molecules, plays a major role in the proteome. Zong et al. present DeepGP, a hybrid deep learning framework based on transformer and graph neural network architectures that accurately predicts tandem mass spectra and retention times of glycopeptides, providing information on glycosylation and glycan structure.
蛋白质糖基化是蛋白质通过聚糖进行的翻译后修饰,在细胞的多种生理和病理功能中发挥着重要作用。糖蛋白组学是在整个蛋白质组范围内研究蛋白质糖基化的方法,它利用液相色谱法和串联质谱法(MS/MS)获得糖基化位点、糖基化水平和聚糖结构的综合信息。然而,目前用于糖蛋白组学的数据库搜索方法往往由于结构决定离子的出现有限而难以确定糖分子结构。虽然光谱搜索方法可以利用片段强度来促进糖肽的结构鉴定,但其应用受到光谱库构建困难的阻碍。在这项工作中,我们提出了基于变换器和图神经网络的混合深度学习框架 DeepGP,用于预测糖肽的 MS/MS 图谱和保留时间。我们采用了两个图神经网络模块,分别用于捕捉支链聚糖结构和预测聚糖离子强度。此外,还采用了预训练策略,以缓解糖蛋白组学数据不足的问题。在多个生物数据集上进行测试后,DeepGP 准确预测了糖肽的 MS/MS 图谱和保留时间,与实验结果非常吻合。在合成和生物数据集上对 DeepGP 进行的全面基准测试验证了它在区分相似聚糖方面的有效性。基于各种诱饵方法,DeepGP 与数据库搜索相结合可以提高糖肽检测灵敏度。我们预计,DeepGP 将激发未来在糖蛋白组学领域的广泛工作。
{"title":"Deep learning prediction of glycopeptide tandem mass spectra powers glycoproteomics","authors":"Yu Zong,&nbsp;Yuxin Wang,&nbsp;Xipeng Qiu,&nbsp;Xuanjing Huang,&nbsp;Liang Qiao","doi":"10.1038/s42256-024-00875-x","DOIUrl":"10.1038/s42256-024-00875-x","url":null,"abstract":"Protein glycosylation, a post-translational modification of proteins by glycans, plays an important role in numerous physiological and pathological cellular functions. Glycoproteomics, the study of protein glycosylation on a proteome-wide scale, utilizes liquid chromatography coupled with tandem mass spectrometry (MS/MS) to get combinational information on glycosylation site, glycosylation level and glycan structure. However, current database searching methods for glycoproteomics often struggle with glycan structure determination due to the limited occurrence of structure-determining ions. Although spectral searching methods can leverage fragment intensity to facilitate the structure identification of glycopeptides, their application is hindered by difficulties in spectral library construction. In this work, we present DeepGP, a hybrid deep learning framework based on transformer and graph neural networks, for the prediction of MS/MS spectra and retention time of glycopeptides. Two graph neural network modules are employed to capture the branched glycan structure and predict glycan ion intensity, respectively. Additionally, a pretraining strategy is implemented to alleviate the insufficiency of glycoproteomics data. Testing on multiple biological datasets, DeepGP accurately predicts MS/MS spectra and retention time of glycopeptides, closely aligning with the experimental results. Comprehensive benchmarking of DeepGP on synthetic and biological datasets validates its effectiveness in distinguishing similar glycans. Based on various decoy methods, DeepGP in combination with database searching can increase glycopeptide detection sensitivity. We anticipate that DeepGP can inspire extensive future work in glycoproteomics. Glycosylation, a prevalent type of post-translational modification of proteins by glycan molecules, plays a major role in the proteome. Zong et al. present DeepGP, a hybrid deep learning framework based on transformer and graph neural network architectures that accurately predicts tandem mass spectra and retention times of glycopeptides, providing information on glycosylation and glycan structure.","PeriodicalId":48533,"journal":{"name":"Nature Machine Intelligence","volume":null,"pages":null},"PeriodicalIF":18.8,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141794616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"计算机科学","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
Nature Machine Intelligence
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