首页 > 最新文献

Frontiers in Neuroinformatics最新文献

英文 中文
Exploring EEG-based motor imagery decoding: a dual approach using spatial features and spectro-spatial Deep Learning model IFNet 探索基于脑电图的运动图像解码:使用空间特征和光谱空间深度学习模型 IFNet 的双重方法
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-29 DOI: 10.3389/fninf.2024.1345425
Javier V. Juan, Rubén Martínez, Eduardo Iáñez, Mario Ortiz, Jesús Tornero, José M. Azorín
IntroductionIn recent years, the decoding of motor imagery (MI) from electroencephalography (EEG) signals has become a focus of research for brain-machine interfaces (BMIs) and neurorehabilitation. However, EEG signals present challenges due to their non-stationarity and the substantial presence of noise commonly found in recordings, making it difficult to design highly effective decoding algorithms. These algorithms are vital for controlling devices in neurorehabilitation tasks, as they activate the patient's motor cortex and contribute to their recovery.MethodsThis study proposes a novel approach for decoding MI during pedalling tasks using EEG signals. A widespread approach is based on feature extraction using Common Spatial Patterns (CSP) followed by a linear discriminant analysis (LDA) as a classifier. The first approach covered in this work aims to investigate the efficacy of a task-discriminative feature extraction method based on CSP filter and LDA classifier. Additionally, the second alternative hypothesis explores the potential of a spectro-spatial Convolutional Neural Network (CNN) to further enhance the performance of the first approach. The proposed CNN architecture combines a preprocessing pipeline based on filter banks in the frequency domain with a convolutional neural network for spectro-temporal and spectro-spatial feature extraction.Results and discussionTo evaluate the approaches and their advantages and disadvantages, EEG data has been recorded from several able-bodied users while pedalling in a cycle ergometer in order to train motor imagery decoding models. The results show levels of accuracy up to 80% in some cases. The CNN approach shows greater accuracy despite higher instability.
引言 近年来,从脑电图(EEG)信号中解码运动图像(MI)已成为脑机接口(BMI)和神经康复的研究重点。然而,脑电信号由于其非稳态性和记录中常见的大量噪声,使得设计高效的解码算法变得十分困难。这些算法对于控制神经康复任务中的设备至关重要,因为它们能激活患者的运动皮层,有助于他们的康复。方法本研究提出了一种利用脑电信号对踩踏任务中的 MI 进行解码的新方法。一种普遍采用的方法是使用通用空间模式(CSP)进行特征提取,然后使用线性判别分析(LDA)作为分类器。本研究涉及的第一种方法旨在研究基于 CSP 过滤器和 LDA 分类器的任务判别特征提取方法的有效性。此外,第二个备选假设探讨了光谱空间卷积神经网络(CNN)的潜力,以进一步提高第一种方法的性能。所提出的 CNN 架构将基于频域滤波器组的预处理管道与用于频谱-时域和频谱-空间特征提取的卷积神经网络相结合。结果与讨论为了评估这些方法及其优缺点,我们记录了几位健全用户在自行车测力计上蹬车时的脑电图数据,以训练运动图像解码模型。结果显示,在某些情况下,准确率高达 80%。尽管不稳定性较高,但 CNN 方法显示出更高的准确性。
{"title":"Exploring EEG-based motor imagery decoding: a dual approach using spatial features and spectro-spatial Deep Learning model IFNet","authors":"Javier V. Juan, Rubén Martínez, Eduardo Iáñez, Mario Ortiz, Jesús Tornero, José M. Azorín","doi":"10.3389/fninf.2024.1345425","DOIUrl":"https://doi.org/10.3389/fninf.2024.1345425","url":null,"abstract":"IntroductionIn recent years, the decoding of motor imagery (MI) from electroencephalography (EEG) signals has become a focus of research for brain-machine interfaces (BMIs) and neurorehabilitation. However, EEG signals present challenges due to their non-stationarity and the substantial presence of noise commonly found in recordings, making it difficult to design highly effective decoding algorithms. These algorithms are vital for controlling devices in neurorehabilitation tasks, as they activate the patient's motor cortex and contribute to their recovery.MethodsThis study proposes a novel approach for decoding MI during pedalling tasks using EEG signals. A widespread approach is based on feature extraction using Common Spatial Patterns (CSP) followed by a linear discriminant analysis (LDA) as a classifier. The first approach covered in this work aims to investigate the efficacy of a task-discriminative feature extraction method based on CSP filter and LDA classifier. Additionally, the second alternative hypothesis explores the potential of a spectro-spatial Convolutional Neural Network (CNN) to further enhance the performance of the first approach. The proposed CNN architecture combines a preprocessing pipeline based on filter banks in the frequency domain with a convolutional neural network for spectro-temporal and spectro-spatial feature extraction.Results and discussionTo evaluate the approaches and their advantages and disadvantages, EEG data has been recorded from several able-bodied users while pedalling in a cycle ergometer in order to train motor imagery decoding models. The results show levels of accuracy up to 80% in some cases. The CNN approach shows greater accuracy despite higher instability.","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140008705","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A scoping review of mathematical models covering Alzheimer's disease progression 涵盖阿尔茨海默病进展的数学模型范围综述
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-28 DOI: 10.3389/fninf.2024.1281656
Seyedadel Moravveji, Nicolas Doyon, Javad Mashreghi, Simon Duchesne

Alzheimer's disease is a complex, multi-factorial, and multi-parametric neurodegenerative etiology. Mathematical models can help understand such a complex problem by providing a way to explore and conceptualize principles, merging biological knowledge with experimental data into a model amenable to simulation and external validation, all without the need for extensive clinical trials. We performed a scoping review of mathematical models describing the onset and evolution of Alzheimer's disease as a result of biophysical factors following the PRISMA standard. Our search strategy applied to the PubMed database yielded 846 entries. After using our exclusion criteria, only 17 studies remained from which we extracted data, which focused on three aspects of mathematical modeling: how authors addressed continuous time (since even when the measurements are punctual, the biological processes underlying Alzheimer's disease evolve continuously), how models were solved, and how the high dimensionality and non-linearity of models were managed. Most articles modeled Alzheimer's disease at the cellular level, operating on a short time scale (e.g., minutes or hours), i.e., the micro view (12/17); the rest considered regional or brain-level processes with longer timescales (e.g., years or decades) (the macro view). Most papers were concerned primarily with amyloid beta (n = 8), few described both amyloid beta and tau proteins (n = 3), while some considered more than these two factors (n = 6). Models used partial differential equations (n = 3), ordinary differential equations (n = 7), and both partial differential equations and ordinary differential equations (n = 3). Some did not specify their mathematical formalism (n = 4). Sensitivity analyses were performed in only a small number of papers (4/17). Overall, we found that only two studies could be considered valid in terms of parameters and conclusions, and two more were partially valid. This puts the majority (n = 13) as being either invalid or with insufficient information to ascertain their status. This was the main finding of our paper, in that serious shortcomings make their results invalid or non-reproducible. These shortcomings come from insufficient methodological description, poor calibration, or the impossibility of experimentally validating or calibrating the model. Those shortcomings should be addressed by future authors to unlock the usefulness of mathematical models in Alzheimer's disease.

阿尔茨海默病是一种复杂、多因素和多参数的神经退行性病变。数学模型可以帮助理解这样一个复杂的问题,它提供了一种探索和概念化原理的方法,将生物知识与实验数据融合成一个可以进行模拟和外部验证的模型,而所有这些都不需要进行大量的临床试验。我们按照 PRISMA 标准,对描述生物物理因素导致阿尔茨海默病发病和演变的数学模型进行了一次范围审查。我们在 PubMed 数据库中采用的搜索策略共搜索到 846 个条目。在使用了排除标准后,只剩下 17 篇研究,我们从中提取了数据,这些数据主要集中在数学建模的三个方面:作者如何处理连续时间(因为即使测量是准时的,阿尔茨海默病的生物过程也在不断演变)、如何求解模型,以及如何处理模型的高维性和非线性。大多数文章从细胞层面对阿尔茨海默病进行建模,在短时间内(如几分钟或几小时)发挥作用,即微观视角(12/17);其余文章考虑了时间尺度较长(如几年或几十年)的区域或大脑层面的过程(宏观视角)。大多数论文主要关注β淀粉样蛋白(8 篇),少数论文同时描述了β淀粉样蛋白和tau蛋白(3 篇),而有些论文考虑的因素超过了这两个因素(6 篇)。模型使用了偏微分方程(3 个)、常微分方程(7 个)以及偏微分方程和常微分方程(3 个)。有些模型没有说明其数学形式(n = 4)。只有少数论文(4/17)进行了敏感性分析。总体而言,我们发现只有两项研究的参数和结论是有效的,另有两项研究的参数和结论是部分有效的。因此,大多数研究(n = 13)要么无效,要么信息不足,无法确定其状态。这也是我们论文的主要发现,即严重的缺陷使其结果无效或不可再现。这些缺陷来自于方法描述不足、校准不佳或无法通过实验验证或校准模型。未来的作者应该解决这些缺陷,以释放数学模型在阿尔茨海默病中的效用。
{"title":"A scoping review of mathematical models covering Alzheimer's disease progression","authors":"Seyedadel Moravveji, Nicolas Doyon, Javad Mashreghi, Simon Duchesne","doi":"10.3389/fninf.2024.1281656","DOIUrl":"https://doi.org/10.3389/fninf.2024.1281656","url":null,"abstract":"<p>Alzheimer's disease is a complex, multi-factorial, and multi-parametric neurodegenerative etiology. Mathematical models can help understand such a complex problem by providing a way to explore and conceptualize principles, merging biological knowledge with experimental data into a model amenable to simulation and external validation, all without the need for extensive clinical trials. We performed a scoping review of mathematical models describing the onset and evolution of Alzheimer's disease as a result of biophysical factors following the PRISMA standard. Our search strategy applied to the PubMed database yielded 846 entries. After using our exclusion criteria, only 17 studies remained from which we extracted data, which focused on three aspects of mathematical modeling: how authors addressed continuous time (since even when the measurements are punctual, the biological processes underlying Alzheimer's disease evolve continuously), how models were solved, and how the high dimensionality and non-linearity of models were managed. Most articles modeled Alzheimer's disease at the cellular level, operating on a short time scale (e.g., minutes or hours), i.e., the micro view (12/17); the rest considered regional or brain-level processes with longer timescales (e.g., years or decades) (the macro view). Most papers were concerned primarily with amyloid beta (<italic>n</italic> = 8), few described both amyloid beta and tau proteins (<italic>n</italic> = 3), while some considered more than these two factors (<italic>n</italic> = 6). Models used partial differential equations (<italic>n</italic> = 3), ordinary differential equations (<italic>n</italic> = 7), and both partial differential equations and ordinary differential equations (<italic>n</italic> = 3). Some did not specify their mathematical formalism (<italic>n</italic> = 4). Sensitivity analyses were performed in only a small number of papers (4/17). Overall, we found that only two studies could be considered valid in terms of parameters and conclusions, and two more were partially valid. This puts the majority (<italic>n</italic> = 13) as being either invalid or with insufficient information to ascertain their status. This was the main finding of our paper, in that serious shortcomings make their results invalid or non-reproducible. These shortcomings come from insufficient methodological description, poor calibration, or the impossibility of experimentally validating or calibrating the model. Those shortcomings should be addressed by future authors to unlock the usefulness of mathematical models in Alzheimer's disease.</p>","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140124651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
suMRak: a multi-tool solution for preclinical brain MRI data analysis suMRak:临床前脑磁共振成像数据分析的多工具解决方案
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-26 DOI: 10.3389/fninf.2024.1358917
Rok Ister, Marko Sternak, Siniša Škokić, Srećko Gajović
Introduction

Magnetic resonance imaging (MRI) is invaluable for understanding brain disorders, but data complexity poses a challenge in experimental research. In this study, we introduce suMRak, a MATLAB application designed for efficient preclinical brain MRI analysis. SuMRak integrates brain segmentation, volumetry, image registration, and parameter map generation into a unified interface, thereby reducing the number of separate tools that researchers may require for straightforward data handling.

Methods and implementation

All functionalities of suMRak are implemented using the MATLAB App Designer and the MATLAB-integrated Python engine. A total of six helper applications were developed alongside the main suMRak interface to allow for a cohesive and streamlined workflow. The brain segmentation strategy was validated by comparing suMRak against manual segmentation and ITK-SNAP, a popular open-source application for biomedical image segmentation.

Results

When compared with the manual segmentation of coronal mouse brain slices, suMRak achieved a high Sørensen–Dice similarity coefficient (0.98 ± 0.01), approaching manual accuracy. Additionally, suMRak exhibited significant improvement (p = 0.03) when compared to ITK-SNAP, particularly for caudally located brain slices. Furthermore, suMRak was capable of effectively analyzing preclinical MRI data obtained in our own studies. Most notably, the results of brain perfusion map registration to T2-weighted images were shown, improving the topographic connection to anatomical areas and enabling further data analysis to better account for the inherent spatial distortions of echoplanar imaging.

Discussion

SuMRak offers efficient MRI data processing of preclinical brain images, enabling researchers' consistency and precision. Notably, the accelerated brain segmentation, achieved through K-means clustering and morphological operations, significantly reduces processing time and allows for easier handling of larger datasets.

导言磁共振成像(MRI)对于了解脑部疾病非常宝贵,但数据的复杂性给实验研究带来了挑战。在本研究中,我们介绍了 suMRak,这是一款专为临床前脑部磁共振成像高效分析而设计的 MATLAB 应用程序。SuMRak 将脑部分割、容积测量、图像配准和参数图生成集成到一个统一的界面中,从而减少了研究人员在直接处理数据时可能需要的独立工具的数量。方法与实现 suMRak 的所有功能都是通过 MATLAB 应用程序设计器和集成 MATLAB 的 Python 引擎实现的。在开发 suMRak 主界面的同时,还开发了总共六个辅助应用程序,以实现连贯、精简的工作流程。通过将 suMRak 与人工分割和 ITK-SNAP(一种用于生物医学图像分割的流行开源应用程序)进行比较,对大脑分割策略进行了验证。结果与小鼠冠状脑切片的人工分割相比,suMRak 实现了较高的 Sørensen-Dice 相似性系数(0.98 ± 0.01),接近人工分割的准确性。此外,与 ITK-SNAP 相比,suMRak 表现出显著的改进(p = 0.03),尤其是在尾部位置的脑片上。此外,suMRak 还能有效分析我们自己的研究中获得的临床前 MRI 数据。最值得注意的是,脑灌注图与 T2 加权图像的配准结果显示,改善了与解剖区域的地形连接,并使进一步的数据分析能够更好地考虑回声平面成像固有的空间失真。值得注意的是,通过 K-means 聚类和形态学操作实现的加速脑部分割大大缩短了处理时间,并能更轻松地处理较大的数据集。
{"title":"suMRak: a multi-tool solution for preclinical brain MRI data analysis","authors":"Rok Ister, Marko Sternak, Siniša Škokić, Srećko Gajović","doi":"10.3389/fninf.2024.1358917","DOIUrl":"https://doi.org/10.3389/fninf.2024.1358917","url":null,"abstract":"<sec><title>Introduction</title><p>Magnetic resonance imaging (MRI) is invaluable for understanding brain disorders, but data complexity poses a challenge in experimental research. In this study, we introduce suMRak, a MATLAB application designed for efficient preclinical brain MRI analysis. SuMRak integrates brain segmentation, volumetry, image registration, and parameter map generation into a unified interface, thereby reducing the number of separate tools that researchers may require for straightforward data handling.</p></sec><sec><title>Methods and implementation</title><p>All functionalities of suMRak are implemented using the MATLAB App Designer and the MATLAB-integrated Python engine. A total of six helper applications were developed alongside the main suMRak interface to allow for a cohesive and streamlined workflow. The brain segmentation strategy was validated by comparing suMRak against manual segmentation and ITK-SNAP, a popular open-source application for biomedical image segmentation.</p></sec><sec><title>Results</title><p>When compared with the manual segmentation of coronal mouse brain slices, suMRak achieved a high Sørensen–Dice similarity coefficient (0.98 ± 0.01), approaching manual accuracy. Additionally, suMRak exhibited significant improvement (<italic>p</italic> = 0.03) when compared to ITK-SNAP, particularly for caudally located brain slices. Furthermore, suMRak was capable of effectively analyzing preclinical MRI data obtained in our own studies. Most notably, the results of brain perfusion map registration to T2-weighted images were shown, improving the topographic connection to anatomical areas and enabling further data analysis to better account for the inherent spatial distortions of echoplanar imaging.</p></sec><sec><title>Discussion</title><p>SuMRak offers efficient MRI data processing of preclinical brain images, enabling researchers' consistency and precision. Notably, the accelerated brain segmentation, achieved through K-means clustering and morphological operations, significantly reduces processing time and allows for easier handling of larger datasets.</p></sec>","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140299170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Accelerating spiking neural network simulations with PymoNNto and PymoNNtorch 利用 PymoNNto 和 PymoNNtorch 加速尖峰神经网络仿真
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-20 DOI: 10.3389/fninf.2024.1331220
Marius Vieth, Ali Rahimi, Ashena Gorgan Mohammadi, Jochen Triesch, Mohammad Ganjtabesh
Spiking neural network simulations are a central tool in Computational Neuroscience, Artificial Intelligence, and Neuromorphic Engineering research. A broad range of simulators and software frameworks for such simulations exist with different target application areas. Among these, PymoNNto is a recent Python-based toolbox for spiking neural network simulations that emphasizes the embedding of custom code in a modular and flexible way. While PymoNNto already supports GPU implementations, its backend relies on NumPy operations. Here we introduce PymoNNtorch, which is natively implemented with PyTorch while retaining PymoNNto's modular design. Furthermore, we demonstrate how changes to the implementations of common network operations in combination with PymoNNtorch's native GPU support can offer speed-up over conventional simulators like NEST, ANNarchy, and Brian 2 in certain situations. Overall, we show how PymoNNto's modular and flexible design in combination with PymoNNtorch's GPU acceleration and optimized indexing operations facilitate research and development of spiking neural networks in the Python programming language.
尖峰神经网络模拟是计算神经科学、人工智能和神经形态工程研究的核心工具。针对不同的目标应用领域,有多种用于此类模拟的模拟器和软件框架。其中,PymoNNto 是最近推出的一款基于 Python 的尖峰神经网络仿真工具箱,它强调以模块化和灵活的方式嵌入自定义代码。虽然 PymoNNto 已经支持 GPU 实现,但其后台依赖于 NumPy 操作。在这里,我们将介绍 PymoNNtorch,它是用 PyTorch 原生实现的,同时保留了 PymoNNto 的模块化设计。此外,我们还展示了如何通过改变常见网络操作的实现方式,结合 PymoNNtorch 的原生 GPU 支持,在某些情况下实现比 NEST、ANNarchy 和 Brian 2 等传统模拟器更快的速度。总之,我们展示了 PymoNNto 的模块化和灵活设计如何与 PymoNNtorch 的 GPU 加速和优化索引操作相结合,促进了 Python 编程语言中尖峰神经网络的研究与开发。
{"title":"Accelerating spiking neural network simulations with PymoNNto and PymoNNtorch","authors":"Marius Vieth, Ali Rahimi, Ashena Gorgan Mohammadi, Jochen Triesch, Mohammad Ganjtabesh","doi":"10.3389/fninf.2024.1331220","DOIUrl":"https://doi.org/10.3389/fninf.2024.1331220","url":null,"abstract":"Spiking neural network simulations are a central tool in Computational Neuroscience, Artificial Intelligence, and Neuromorphic Engineering research. A broad range of simulators and software frameworks for such simulations exist with different target application areas. Among these, PymoNNto is a recent Python-based toolbox for spiking neural network simulations that emphasizes the embedding of custom code in a modular and flexible way. While PymoNNto already supports GPU implementations, its backend relies on NumPy operations. Here we introduce PymoNNtorch, which is natively implemented with PyTorch while retaining PymoNNto's modular design. Furthermore, we demonstrate how changes to the implementations of common network operations in combination with PymoNNtorch's native GPU support can offer speed-up over conventional simulators like NEST, ANNarchy, and Brian 2 in certain situations. Overall, we show how PymoNNto's modular and flexible design in combination with PymoNNtorch's GPU acceleration and optimized indexing operations facilitate research and development of spiking neural networks in the Python programming language.","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139918181","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Empirical comparison of deep learning models for fNIRS pain decoding 用于 fNIRS 疼痛解码的深度学习模型的经验比较
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-14 DOI: 10.3389/fninf.2024.1320189
Raul Fernandez Rojas, Calvin Joseph, Ghazal Bargshady, Keng-Liang Ou
IntroductionPain assessment is extremely important in patients unable to communicate and it is often done by clinical judgement. However, assessing pain using observable indicators can be challenging for clinicians due to the subjective perceptions, individual differences in pain expression, and potential confounding factors. Therefore, the need for an objective pain assessment method that can assist medical practitioners. Functional near-infrared spectroscopy (fNIRS) has shown promising results to assess the neural function in response of nociception and pain. Previous studies have explored the use of machine learning with hand-crafted features in the assessment of pain.MethodsIn this study, we aim to expand previous studies by exploring the use of deep learning models Convolutional Neural Network (CNN), Long Short-Term Memory (LSTM), and (CNN-LSTM) to automatically extract features from fNIRS data and by comparing these with classical machine learning models using hand-crafted features.ResultsThe results showed that the deep learning models exhibited favourable results in the identification of different types of pain in our experiment using only fNIRS input data. The combination of CNN and LSTM in a hybrid model (CNN-LSTM) exhibited the highest performance (accuracy = 91.2%) in our problem setting. Statistical analysis using one-way ANOVA with Tukey's (post-hoc) test performed on accuracies showed that the deep learning models significantly improved accuracy performance as compared to the baseline models.DiscussionOverall, deep learning models showed their potential to learn features automatically without relying on manually-extracted features and the CNN-LSTM model could be used as a possible method of assessment of pain in non-verbal patients. Future research is needed to evaluate the generalisation of this method of pain assessment on independent populations and in real-life scenarios.
导言:疼痛评估对于无法交流的患者极为重要,通常需要通过临床判断来完成。然而,由于主观感受、疼痛表现的个体差异以及潜在的混杂因素,使用可观察的指标来评估疼痛对临床医生来说具有挑战性。因此,需要一种客观的疼痛评估方法来帮助医生。功能性近红外光谱(fNIRS)在评估神经功能对痛觉和疼痛的反应方面显示出良好的效果。方法在本研究中,我们旨在扩展之前的研究,探索使用深度学习模型卷积神经网络(CNN)、长短期记忆(LSTM)和(CNN-LSTM)从 fNIRS 数据中自动提取特征,并将其与使用手工创建特征的经典机器学习模型进行比较。结果结果表明,在仅使用 fNIRS 输入数据的实验中,深度学习模型在识别不同类型疼痛方面表现出了良好的效果。在我们的问题设置中,CNN 和 LSTM 的混合模型(CNN-LSTM)表现出了最高的性能(准确率 = 91.2%)。使用单向方差分析和 Tukey's(事后)检验对准确率进行的统计分析表明,与基线模型相比,深度学习模型显著提高了准确率。未来的研究需要评估这种疼痛评估方法在独立人群和现实生活场景中的通用性。
{"title":"Empirical comparison of deep learning models for fNIRS pain decoding","authors":"Raul Fernandez Rojas, Calvin Joseph, Ghazal Bargshady, Keng-Liang Ou","doi":"10.3389/fninf.2024.1320189","DOIUrl":"https://doi.org/10.3389/fninf.2024.1320189","url":null,"abstract":"IntroductionPain assessment is extremely important in patients unable to communicate and it is often done by clinical judgement. However, assessing pain using observable indicators can be challenging for clinicians due to the subjective perceptions, individual differences in pain expression, and potential confounding factors. Therefore, the need for an objective pain assessment method that can assist medical practitioners. Functional near-infrared spectroscopy (fNIRS) has shown promising results to assess the neural function in response of nociception and pain. Previous studies have explored the use of machine learning with hand-crafted features in the assessment of pain.MethodsIn this study, we aim to expand previous studies by exploring the use of deep learning models Convolutional Neural Network (CNN), Long Short-Term Memory (LSTM), and (CNN-LSTM) to automatically extract features from fNIRS data and by comparing these with classical machine learning models using hand-crafted features.ResultsThe results showed that the deep learning models exhibited favourable results in the identification of different types of pain in our experiment using only fNIRS input data. The combination of CNN and LSTM in a hybrid model (CNN-LSTM) exhibited the highest performance (accuracy = 91.2%) in our problem setting. Statistical analysis using one-way ANOVA with Tukey's (<jats:italic>post-hoc</jats:italic>) test performed on accuracies showed that the deep learning models significantly improved accuracy performance as compared to the baseline models.DiscussionOverall, deep learning models showed their potential to learn features automatically without relying on manually-extracted features and the CNN-LSTM model could be used as a possible method of assessment of pain in non-verbal patients. Future research is needed to evaluate the generalisation of this method of pain assessment on independent populations and in real-life scenarios.","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139768382","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multiscale co-simulation design pattern for neuroscience applications 神经科学应用的多尺度协同仿真设计模式
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-12 DOI: 10.3389/fninf.2024.1156683
Lionel Kusch, Sandra Diaz-Pier, Wouter Klijn, Kim Sontheimer, Christophe Bernard, Abigail Morrison, Viktor Jirsa
Integration of information across heterogeneous sources creates added scientific value. Interoperability of data, tools and models is, however, difficult to accomplish across spatial and temporal scales. Here we introduce the toolbox Parallel Co-Simulation, which enables the interoperation of simulators operating at different scales. We provide a software science co-design pattern and illustrate its functioning along a neuroscience example, in which individual regions of interest are simulated on the cellular level allowing us to study detailed mechanisms, while the remaining network is efficiently simulated on the population level. A workflow is illustrated for the use case of The Virtual Brain and NEST, in which the CA1 region of the cellular-level hippocampus of the mouse is embedded into a full brain network involving micro and macro electrode recordings. This new tool allows integrating knowledge across scales in the same simulation framework and validating them against multiscale experiments, thereby largely widening the explanatory power of computational models.
整合不同来源的信息可以创造更多的科学价值。然而,跨时空尺度的数据、工具和模型的互操作性很难实现。在此,我们介绍了并行协同仿真工具箱,该工具箱可实现不同规模的仿真器之间的互操作。我们提供了一种软件科学协同设计模式,并以神经科学为例说明了它的功能,其中单个感兴趣的区域在细胞水平上进行模拟,使我们能够研究详细的机制,而其余网络则在群体水平上进行有效模拟。图中展示了虚拟大脑和 NEST 使用案例的工作流程,其中小鼠细胞级海马 CA1 区域被嵌入到涉及微观和宏观电极记录的完整大脑网络中。这一新工具允许在同一模拟框架中整合跨尺度知识,并根据多尺度实验对其进行验证,从而在很大程度上拓宽了计算模型的解释能力。
{"title":"Multiscale co-simulation design pattern for neuroscience applications","authors":"Lionel Kusch, Sandra Diaz-Pier, Wouter Klijn, Kim Sontheimer, Christophe Bernard, Abigail Morrison, Viktor Jirsa","doi":"10.3389/fninf.2024.1156683","DOIUrl":"https://doi.org/10.3389/fninf.2024.1156683","url":null,"abstract":"Integration of information across heterogeneous sources creates added scientific value. Interoperability of data, tools and models is, however, difficult to accomplish across spatial and temporal scales. Here we introduce the toolbox Parallel Co-Simulation, which enables the interoperation of simulators operating at different scales. We provide a software science co-design pattern and illustrate its functioning along a neuroscience example, in which individual regions of interest are simulated on the cellular level allowing us to study detailed mechanisms, while the remaining network is efficiently simulated on the population level. A workflow is illustrated for the use case of The Virtual Brain and NEST, in which the CA1 region of the cellular-level hippocampus of the mouse is embedded into a full brain network involving micro and macro electrode recordings. This new tool allows integrating knowledge across scales in the same simulation framework and validating them against multiscale experiments, thereby largely widening the explanatory power of computational models.","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139768383","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Locare workflow: representing neuroscience data locations as geometric objects in 3D brain atlases Locare 工作流程:在三维脑图谱中将神经科学数据位置表示为几何对象
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-09 DOI: 10.3389/fninf.2024.1284107
Camilla H. Blixhavn, Ingrid Reiten, Heidi Kleven, Martin Øvsthus, Sharon C. Yates, Ulrike Schlegel, Maja A. Puchades, Oliver Schmid, Jan G. Bjaalie, Ingvild E. Bjerke, Trygve B. Leergaard
Neuroscientists employ a range of methods and generate increasing amounts of data describing brain structure and function. The anatomical locations from which observations or measurements originate represent a common context for data interpretation, and a starting point for identifying data of interest. However, the multimodality and abundance of brain data pose a challenge for efforts to organize, integrate, and analyze data based on anatomical locations. While structured metadata allow faceted data queries, different types of data are not easily represented in a standardized and machine-readable way that allow comparison, analysis, and queries related to anatomical relevance. To this end, three-dimensional (3D) digital brain atlases provide frameworks in which disparate multimodal and multilevel neuroscience data can be spatially represented. We propose to represent the locations of different neuroscience data as geometric objects in 3D brain atlases. Such geometric objects can be specified in a standardized file format and stored as location metadata for use with different computational tools. We here present the Locare workflow developed for defining the anatomical location of data elements from rodent brains as geometric objects. We demonstrate how the workflow can be used to define geometric objects representing multimodal and multilevel experimental neuroscience in rat or mouse brain atlases. We further propose a collection of JSON schemas (LocareJSON) for specifying geometric objects by atlas coordinates, suitable as a starting point for co-visualization of different data in an anatomical context and for enabling spatial data queries.
神经科学家采用一系列方法,产生了越来越多描述大脑结构和功能的数据。观测或测量的解剖位置是解释数据的共同背景,也是识别相关数据的起点。然而,大脑数据的多模态性和丰富性给根据解剖位置组织、整合和分析数据的工作带来了挑战。虽然结构化元数据允许分面数据查询,但不同类型的数据不易以标准化和机器可读的方式表示,因此无法进行比较、分析和与解剖相关性有关的查询。为此,三维(3D)数字脑图谱提供了一个框架,可在其中对不同的多模态和多层次神经科学数据进行空间表示。我们建议在三维脑图谱中将不同神经科学数据的位置表示为几何对象。这些几何对象可以用标准化文件格式指定,并存储为位置元数据,供不同的计算工具使用。我们在此介绍为将啮齿类动物大脑中的数据元素定义为几何对象而开发的 Locare 工作流程。我们演示了如何使用该工作流程定义几何对象,以表示大鼠或小鼠脑图谱中的多模态和多层次实验神经科学。我们进一步提出了一个 JSON 方案集(LocareJSON),用于通过图集坐标指定几何对象,适合作为在解剖学背景下对不同数据进行协同可视化和实现空间数据查询的起点。
{"title":"The Locare workflow: representing neuroscience data locations as geometric objects in 3D brain atlases","authors":"Camilla H. Blixhavn, Ingrid Reiten, Heidi Kleven, Martin Øvsthus, Sharon C. Yates, Ulrike Schlegel, Maja A. Puchades, Oliver Schmid, Jan G. Bjaalie, Ingvild E. Bjerke, Trygve B. Leergaard","doi":"10.3389/fninf.2024.1284107","DOIUrl":"https://doi.org/10.3389/fninf.2024.1284107","url":null,"abstract":"Neuroscientists employ a range of methods and generate increasing amounts of data describing brain structure and function. The anatomical locations from which observations or measurements originate represent a common context for data interpretation, and a starting point for identifying data of interest. However, the multimodality and abundance of brain data pose a challenge for efforts to organize, integrate, and analyze data based on anatomical locations. While structured metadata allow faceted data queries, different types of data are not easily represented in a standardized and machine-readable way that allow comparison, analysis, and queries related to anatomical relevance. To this end, three-dimensional (3D) digital brain atlases provide frameworks in which disparate multimodal and multilevel neuroscience data can be spatially represented. We propose to represent the locations of different neuroscience data as geometric objects in 3D brain atlases. Such geometric objects can be specified in a standardized file format and stored as location metadata for use with different computational tools. We here present the Locare workflow developed for defining the anatomical location of data elements from rodent brains as geometric objects. We demonstrate how the workflow can be used to define geometric objects representing multimodal and multilevel experimental neuroscience in rat or mouse brain atlases. We further propose a collection of JSON schemas (LocareJSON) for specifying geometric objects by atlas coordinates, suitable as a starting point for co-visualization of different data in an anatomical context and for enabling spatial data queries.","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139768385","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In silico analyses of the involvement of GPR55, CB1R and TRPV1: response to THC, contribution to temporal lobe epilepsy, structural modeling and updated evolution 关于 GPR55、CB1R 和 TRPV1 参与情况的硅学分析:对 THC 的反应、对颞叶癫痫的贡献、结构建模和最新演化
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-07 DOI: 10.3389/fninf.2024.1294939
Amy L. Cherry, Michael J. Wheeler, Karolina Mathisova, Mathieu Di Miceli
IntroductionThe endocannabinoid (eCB) system is named after the discovery that endogenous cannabinoids bind to the same receptors as the phytochemical compounds found in Cannabis. While endogenous cannabinoids include anandamide (AEA) and 2-arachidonoylglycerol (2-AG), exogenous phytocannabinoids include Δ-9 tetrahydrocannabinol (THC) and cannabidiol (CBD). These compounds finely tune neurotransmission following synapse activation, via retrograde signaling that activates cannabinoid receptor 1 (CB1R) and/or transient receptor potential cation channel subfamily V member 1 (TRPV1). Recently, the eCB system has been linked to several neurological diseases, such as neuro-ocular abnormalities, pain insensitivity, migraine, epilepsy, addiction and neurodevelopmental disorders. In the current study, we aim to: (i) highlight a potential link between the eCB system and neurological disorders, (ii) assess if THC exposure alters the expression of eCB-related genes, and (iii) identify evolutionary-conserved residues in CB1R or TRPV1 in light of their function.MethodsTo address this, we used several bioinformatic approaches, such as transcriptomic (Gene Expression Omnibus), protein–protein (STRING), phylogenic (BLASTP, MEGA) and structural (Phyre2, AutoDock, Vina, PyMol) analyzes.ResultsUsing RNA sequencing datasets, we did not observe any dysregulation of eCB-related transcripts in major depressive disorders, bipolar disorder or schizophrenia in the anterior cingulate cortex, nucleus accumbens or dorsolateral striatum. Following in vivo THC exposure in adolescent mice, GPR55 was significantly upregulated in neurons from the ventral tegmental area, while other transcripts involved in the eCB system were not affected by THC exposure. Our results also suggest that THC likely induces neuroinflammation following in vitro application on mice microglia. Significant downregulation of TPRV1 occurred in the hippocampi of mice in which a model of temporal lobe epilepsy was induced, confirming previous observations. In addition, several transcriptomic dysregulations were observed in neurons of both epileptic mice and humans, which included transcripts involved in neuronal death. When scanning known interactions for transcripts involved in the eCB system (n = 12), we observed branching between the eCB system and neurophysiology, including proteins involved in the dopaminergic system. Our protein phylogenic analyzes revealed that CB1R forms a clade with CB2R, which is distinct from related paralogues such as sphingosine-1-phosphate, receptors, lysophosphatidic acid receptors and melanocortin receptors. As expected, several conserved residues were identified, which are crucial for CB1R receptor function. The anandamide-binding pocket seems to have appeared later in evolution. Similar results were observed for TRPV1, with conserved residues involved in receptor activa
导言内源性大麻素(eCB)系统因发现内源性大麻素与大麻中的植物化学物质结合到相同的受体而得名。内源性大麻素包括anandamide(AEA)和2-arachidonoylglycerol(2-AG),而外源性植物大麻素包括Δ-9 tetrahydrocannabinol(THC)和大麻二酚(CBD)。这些化合物通过逆行信号激活大麻素受体 1(CB1R)和/或瞬时受体电位阳离子通道 V 亚家族成员 1(TRPV1),从而在突触激活后对神经传递进行微调。最近,eCB 系统与多种神经系统疾病有关,如神经-眼部异常、疼痛不敏感、偏头痛、癫痫、成瘾和神经发育障碍。本研究旨在(i) 强调 eCB 系统与神经系统疾病之间的潜在联系;(ii) 评估暴露于 THC 是否会改变 eCB 相关基因的表达;(iii) 根据 CB1R 或 TRPV1 的功能确定其进化保守残基。方法为了解决这个问题,我们使用了多种生物信息学方法,如转录组(基因表达总库)、蛋白质-蛋白质(STRING)、系统发生(BLASTP、MEGA)和结构(Phyre2、AutoDock、Vina、PyMol)分析。结果通过RNA测序数据集,我们没有观察到重度抑郁症、双相情感障碍或精神分裂症患者前扣带回皮层、伏隔核或背外侧纹状体中eCB相关转录本的失调。青少年小鼠体内暴露 THC 后,腹侧被盖区神经元中的 GPR55 显著上调,而其他涉及 eCB 系统的转录本则不受 THC 暴露的影响。我们的研究结果还表明,体外应用 THC 可能会诱发小鼠小胶质细胞的神经炎症。在诱导颞叶癫痫模型的小鼠海马中,TPRV1发生了显著的下调,这证实了之前的观察结果。此外,在癫痫小鼠和人类的神经元中都观察到了几种转录组失调,其中包括涉及神经元死亡的转录本。在扫描涉及 eCB 系统(n = 12)的转录本的已知相互作用时,我们观察到 eCB 系统和神经生理学之间的分支,包括涉及多巴胺能系统的蛋白质。我们的蛋白质系统发育分析表明,CB1R 与 CB2R 形成一个支系,与鞘氨醇-1-磷酸、受体、溶血磷脂酸受体和黑皮质素受体等相关旁系亲属不同。不出所料,我们发现了几个保守残基,它们对 CB1R 受体的功能至关重要。安乃近结合口袋似乎在进化过程中出现较晚。结论目前的研究发现,暴露于 THC 后神经元中 GPR55 上调,而 TRPV1 在颞叶癫痫中下调。由于我们采用了二次分析,因此在解释本研究结果时应谨慎。CB1R 和 TRPV1 的共同祖先是在 4.5 亿年前的奥陶纪晚期从无颌脊椎动物中分化出来的。我们发现了介导关键受体功能的保守残基。
{"title":"In silico analyses of the involvement of GPR55, CB1R and TRPV1: response to THC, contribution to temporal lobe epilepsy, structural modeling and updated evolution","authors":"Amy L. Cherry, Michael J. Wheeler, Karolina Mathisova, Mathieu Di Miceli","doi":"10.3389/fninf.2024.1294939","DOIUrl":"https://doi.org/10.3389/fninf.2024.1294939","url":null,"abstract":"IntroductionThe endocannabinoid (eCB) system is named after the discovery that endogenous cannabinoids bind to the same receptors as the phytochemical compounds found in Cannabis. While endogenous cannabinoids include anandamide (AEA) and 2-arachidonoylglycerol (2-AG), exogenous phytocannabinoids include Δ-9 tetrahydrocannabinol (THC) and cannabidiol (CBD). These compounds finely tune neurotransmission following synapse activation, via retrograde signaling that activates cannabinoid receptor 1 (CB1R) and/or transient receptor potential cation channel subfamily V member 1 (TRPV1). Recently, the eCB system has been linked to several neurological diseases, such as neuro-ocular abnormalities, pain insensitivity, migraine, epilepsy, addiction and neurodevelopmental disorders. In the current study, we aim to: (i) highlight a potential link between the eCB system and neurological disorders, (ii) assess if THC exposure alters the expression of eCB-related genes, and (iii) identify evolutionary-conserved residues in CB1R or TRPV1 in light of their function.MethodsTo address this, we used several bioinformatic approaches, such as transcriptomic (Gene Expression Omnibus), protein–protein (STRING), phylogenic (BLASTP, MEGA) and structural (Phyre2, AutoDock, Vina, PyMol) analyzes.ResultsUsing RNA sequencing datasets, we did not observe any dysregulation of eCB-related transcripts in major depressive disorders, bipolar disorder or schizophrenia in the anterior cingulate cortex, nucleus accumbens or dorsolateral striatum. Following <jats:italic>in vivo</jats:italic> THC exposure in adolescent mice, <jats:italic>GPR55</jats:italic> was significantly upregulated in neurons from the ventral tegmental area, while other transcripts involved in the eCB system were not affected by THC exposure. Our results also suggest that THC likely induces neuroinflammation following <jats:italic>in vitro</jats:italic> application on mice microglia. Significant downregulation of <jats:italic>TPRV1</jats:italic> occurred in the hippocampi of mice in which a model of temporal lobe epilepsy was induced, confirming previous observations. In addition, several transcriptomic dysregulations were observed in neurons of both epileptic mice and humans, which included transcripts involved in neuronal death. When scanning known interactions for transcripts involved in the eCB system (n = 12), we observed branching between the eCB system and neurophysiology, including proteins involved in the dopaminergic system. Our protein phylogenic analyzes revealed that CB1R forms a clade with CB2R, which is distinct from related paralogues such as sphingosine-1-phosphate, receptors, lysophosphatidic acid receptors and melanocortin receptors. As expected, several conserved residues were identified, which are crucial for CB1R receptor function. The anandamide-binding pocket seems to have appeared later in evolution. Similar results were observed for TRPV1, with conserved residues involved in receptor activa","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139768384","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enabling uncertainty estimation in neural networks through weight perturbation for improved Alzheimer's disease classification 通过权重扰动实现神经网络中的不确定性估计,从而改进阿尔茨海默病分类
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-06 DOI: 10.3389/fninf.2024.1346723
Matteo Ferrante, Tommaso Boccato, Nicola Toschi
BackgroundThe willingness to trust predictions formulated by automatic algorithms is key in a wide range of domains. However, a vast number of deep architectures are only able to formulate predictions without associated uncertainty.PurposeIn this study, we propose a method to convert a standard neural network into a Bayesian neural network and estimate the variability of predictions by sampling different networks similar to the original one at each forward pass.MethodsWe combine our method with a tunable rejection-based approach that employs only the fraction of the data, i.e., the share that the model can classify with an uncertainty below a user-set threshold. We test our model in a large cohort of brain images from patients with Alzheimer's disease and healthy controls, discriminating the former and latter classes based on morphometric images exclusively.ResultsWe demonstrate how combining estimated uncertainty with a rejection-based approach increases classification accuracy from 0.86 to 0.95 while retaining 75% of the test set. In addition, the model can select the cases to be recommended for, e.g., expert human evaluation due to excessive uncertainty. Importantly, our framework circumvents additional workload during the training phase by using our network “turned into Bayesian” to implicitly investigate the loss landscape in the neighborhood of each test sample in order to determine the reliability of the predictions.ConclusionWe believe that being able to estimate the uncertainty of a prediction, along with tools that can modulate the behavior of the network to a degree of confidence that the user is informed about (and comfortable with), can represent a crucial step in the direction of user compliance and easier integration of deep learning tools into everyday tasks currently performed by human operators.
背景对自动算法所做预测的信任度是众多领域的关键。在本研究中,我们提出了一种将标准神经网络转换为贝叶斯神经网络的方法,并通过在每次前向传递时采样与原始网络类似的不同网络来估计预测的可变性。方法我们将我们的方法与基于可调剔除的方法相结合,该方法仅采用数据的一部分,即模型可以分类的不确定性低于用户设置阈值的份额。我们在一大批阿尔茨海默病患者和健康对照者的大脑图像中测试了我们的模型,并完全根据形态计量图像区分了前者和后者。此外,由于不确定性过大,该模型还可以选择推荐的案例,例如进行专家人工评估。重要的是,我们的框架通过使用 "变成贝叶斯 "的网络来隐式地调查每个测试样本附近的损失情况,从而确定预测的可靠性,从而避免了训练阶段的额外工作量。
{"title":"Enabling uncertainty estimation in neural networks through weight perturbation for improved Alzheimer's disease classification","authors":"Matteo Ferrante, Tommaso Boccato, Nicola Toschi","doi":"10.3389/fninf.2024.1346723","DOIUrl":"https://doi.org/10.3389/fninf.2024.1346723","url":null,"abstract":"BackgroundThe willingness to trust predictions formulated by automatic algorithms is key in a wide range of domains. However, a vast number of deep architectures are only able to formulate predictions without associated uncertainty.PurposeIn this study, we propose a method to convert a standard neural network into a Bayesian neural network and estimate the variability of predictions by sampling different networks similar to the original one at each forward pass.MethodsWe combine our method with a tunable rejection-based approach that employs only the fraction of the data, i.e., the share that the model can classify with an uncertainty below a user-set threshold. We test our model in a large cohort of brain images from patients with Alzheimer's disease and healthy controls, discriminating the former and latter classes based on morphometric images exclusively.ResultsWe demonstrate how combining estimated uncertainty with a rejection-based approach increases classification accuracy from 0.86 to 0.95 while retaining 75% of the test set. In addition, the model can select the cases to be recommended for, e.g., expert human evaluation due to excessive uncertainty. Importantly, our framework circumvents additional workload during the training phase by using our network “turned into Bayesian” to implicitly investigate the loss landscape in the neighborhood of each test sample in order to determine the reliability of the predictions.ConclusionWe believe that being able to estimate the uncertainty of a prediction, along with tools that can modulate the behavior of the network to a degree of confidence that the user is informed about (and comfortable with), can represent a crucial step in the direction of user compliance and easier integration of deep learning tools into everyday tasks currently performed by human operators.","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139768352","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Retraction: NeuroSuites: an online platform for running neuroscience, statistical, and machine learning tools. 撤回:NeuroSuites:运行神经科学、统计和机器学习工具的在线平台。
IF 3.5 4区 医学 Q1 Neuroscience Pub Date : 2024-02-05 eCollection Date: 2024-01-01 DOI: 10.3389/fninf.2024.1376953

[This retracts the article DOI: 10.3389/fninf.2023.1092967.].

[本文撤回了文章 DOI:10.3389/fninf.2023.1092967.]。
{"title":"Retraction: NeuroSuites: an online platform for running neuroscience, statistical, and machine learning tools.","authors":"","doi":"10.3389/fninf.2024.1376953","DOIUrl":"https://doi.org/10.3389/fninf.2024.1376953","url":null,"abstract":"<p><p>[This retracts the article DOI: 10.3389/fninf.2023.1092967.].</p>","PeriodicalId":12462,"journal":{"name":"Frontiers in Neuroinformatics","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10876048/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139905491","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Frontiers in Neuroinformatics
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