Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Knowl Inf Syst ; 65(7): 3099-3124, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39131594

RESUMO

The complex systems in the real-world are commonly associated with multiple types of objects and relations, and heterogeneous graphs are ubiquitous data structures that can inherently represent multi-modal interactions between objects. Generating high-quality heterogeneous graphs allows us to understand the implicit distribution of heterogeneous graphs and provides benchmarks for downstream heterogeneous representation learning tasks. Existing works are limited to either merely generating the graph topology with neglecting local semantic information or only generating the graph without preserving the higher-order structural information and the global heterogeneous distribution in generated graphs. To this end, we formulate a general, end-to-end framework - HGEN for generating novel heterogeneous graphs with a newly proposed heterogeneous walk generator. On top of HGEN, we further develop a network motif generator to better characterize the higher-order structural distribution. A novel heterogeneous graph assembler is further developed to adaptively assemble novel heterogeneous graphs from the generated heterogeneous walks and motifs in a stratified manner. The extended model is proven to preserve the local semantic and heterogeneous global distribution of observed graphs with the theoretical guarantee. Lastly, comprehensive experiments on both synthetic and real-world practical datasets demonstrate the power and efficiency of the proposed method.

2.
IEEE Trans Knowl Data Eng ; 35(10): 10871-10883, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38389564

RESUMO

Recent years have witnessed the rapid development of concept map generation techniques due to their advantages in providing well-structured summarization of knowledge from free texts. Traditional unsupervised methods do not generate task-oriented concept maps, whereas deep generative models require large amounts of training data. In this work, we present GT-D2G (Graph Translation-based Document To Graph), an automatic concept map generation framework that leverages generalized NLP pipelines to derive semantic-rich initial graphs, and translates them into more concise structures under the weak supervision of downstream task labels. The concept maps generated by GT-D2G can provide interpretable summarization of structured knowledge for the input texts, which are demonstrated through human evaluation and case studies on three real-world corpora. Further experiments on the downstream task of document classification show that GT-D2G beats other concept map generation methods. Moreover, we specifically validate the labeling efficiency of GT-D2G in the label-efficient learning setting and the flexibility of generated graph sizes in controlled hyper-parameter studies.

3.
IEEE Trans Knowl Data Eng ; 34(10): 4854-4873, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37915376

RESUMO

Since real-world objects and their interactions are often multi-modal and multi-typed, heterogeneous networks have been widely used as a more powerful, realistic, and generic superclass of traditional homogeneous networks (graphs). Meanwhile, representation learning (a.k.a. embedding) has recently been intensively studied and shown effective for various network mining and analytical tasks. In this work, we aim to provide a unified framework to deeply summarize and evaluate existing research on heterogeneous network embedding (HNE), which includes but goes beyond a normal survey. Since there has already been a broad body of HNE algorithms, as the first contribution of this work, we provide a generic paradigm for the systematic categorization and analysis over the merits of various existing HNE algorithms. Moreover, existing HNE algorithms, though mostly claimed generic, are often evaluated on different datasets. Understandable due to the application favor of HNE, such indirect comparisons largely hinder the proper attribution of improved task performance towards effective data preprocessing and novel technical design, especially considering the various ways possible to construct a heterogeneous network from real-world application data. Therefore, as the second contribution, we create four benchmark datasets with various properties regarding scale, structure, attribute/label availability, and etc. from different sources, towards handy and fair evaluations of HNE algorithms. As the third contribution, we carefully refactor and amend the implementations and create friendly interfaces for 13 popular HNE algorithms, and provide all-around comparisons among them over multiple tasks and experimental settings. By putting all existing HNE algorithms under a unified framework, we aim to provide a universal reference and guideline for the understanding and development of HNE algorithms. Meanwhile, by open-sourcing all data and code, we envision to serve the community with an ready-to-use benchmark platform to test and compare the performance of existing and future HNE algorithms (https://github.com/yangji9181/HNE).

4.
Interdiscip Sci ; 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39325266

RESUMO

Computational drug repositioning, through predicting drug-disease associations (DDA), offers significant potential for discovering new drug indications. Current methods incorporate graph neural networks (GNN) on drug-disease heterogeneous networks to predict DDAs, achieving notable performances compared to traditional machine learning and matrix factorization approaches. However, these methods depend heavily on network topology, hampered by incomplete and noisy network data, and overlook the wealth of biomedical knowledge available. Correspondingly, large language models (LLMs) excel in graph search and relational reasoning, which can possibly enhance the integration of comprehensive biomedical knowledge into drug and disease profiles. In this study, we first investigate the contribution of LLM-inferred knowledge representation in drug repositioning and DDA prediction. A zero-shot prompting template was designed for LLM to extract high-quality knowledge descriptions for drug and disease entities, followed by embedding generation from language models to transform the discrete text to continual numerical representation. Then, we proposed LLM-DDA with three different model architectures (LLM-DDANode Feat, LLM-DDADual GNN, LLM-DDAGNN-AE) to investigate the best fusion mode for LLM-based embeddings. Extensive experiments on four DDA benchmarks show that, LLM-DDAGNN-AE achieved the optimal performance compared to 11 baselines with the overall relative improvement in AUPR of 23.22%, F1-Score of 17.20%, and precision of 25.35%. Meanwhile, selected case studies of involving Prednisone and Allergic Rhinitis highlighted the model's capability to identify reliable DDAs and knowledge descriptions, supported by existing literature. This study showcases the utility of LLMs in drug repositioning with its generality and applicability in other biomedical relation prediction tasks.

5.
Pac Symp Biocomput ; 29: 53-64, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38160269

RESUMO

Functional brain networks represent dynamic and complex interactions among anatomical regions of interest (ROIs), providing crucial clinical insights for neural pattern discovery and disorder diagnosis. In recent years, graph neural networks (GNNs) have proven immense success and effectiveness in analyzing structured network data. However, due to the high complexity of data acquisition, resulting in limited training resources of neuroimaging data, GNNs, like all deep learning models, suffer from overfitting. Moreover, their capability to capture useful neural patterns for downstream prediction is also adversely affected. To address such challenge, this study proposes BrainSTEAM, an integrated framework featuring a spatio-temporal module that consists of an EdgeConv GNN model, an autoencoder network, and a Mixup strategy. In particular, the spatio-temporal module aims to dynamically segment the time series signals of the ROI features for each subject into chunked sequences. We leverage each sequence to construct correlation networks, thereby increasing the training data. Additionally, we employ the EdgeConv GNN to capture ROI connectivity structures, an autoencoder for data denoising, and mixup for enhancing model training through linear data augmentation. We evaluate our framework on two real-world neuroimaging datasets, ABIDE for Autism prediction and HCP for gender prediction. Extensive experiments demonstrate the superiority and robustness of BrainSTEAM when compared to a variety of existing models, showcasing the strong potential of our proposed mechanisms in generalizing to other studies for connectome-based fMRI analysis.


Assuntos
Conectoma , Imageamento por Ressonância Magnética , Humanos , Biologia Computacional , Neuroimagem , Encéfalo/diagnóstico por imagem
6.
Pac Symp Biocomput ; 29: 214-225, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38160281

RESUMO

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


Assuntos
Conectoma , Humanos , Biologia Computacional , Encéfalo/diagnóstico por imagem , Redes Neurais de Computação , Neuroimagem
7.
Comput Struct Biotechnol J ; 23: 3020-3029, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39171252

RESUMO

Colorectal cancer (CRC) is the third most diagnosed cancer and the second deadliest cancer worldwide representing a major public health problem. In recent years, increasing evidence has shown that microRNA (miRNA) can control the expression of targeted human messenger RNA (mRNA) by reducing their abundance or translation, acting as oncogenes or tumor suppressors in various cancers, including CRC. Due to the significant up-regulation of oncogenic miRNAs in CRC, elucidating the underlying mechanism and identifying dysregulated miRNA targets may provide a basis for improving current therapeutic interventions. In this paper, we proposed Gra-CRC-miRTar, a pre-trained nucleotide-to-graph neural network framework, for identifying potential miRNA targets in CRC. Different from previous studies, we constructed two pre-trained models to encode RNA sequences and transformed them into de Bruijn graphs. We employed different graph neural networks to learn the latent representations. The embeddings generated from de Bruijn graphs were then fed into a Multilayer Perceptron (MLP) to perform the prediction tasks. Our extensive experiments show that Gra-CRC-miRTar achieves better performance than other deep learning algorithms and existing predictors. In addition, our analyses also successfully revealed 172 out of 201 functional interactions through experimentally validated miRNA-mRNA pairs in CRC. Collectively, our effort provides an accurate and efficient framework to identify potential miRNA targets in CRC, which can also be used to reveal miRNA target interactions in other malignancies, facilitating the development of novel therapeutics. The Gra-CRC-miRTar web server can be found at: http://gra-crc-mirtar.com/.

8.
bioRxiv ; 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38659732

RESUMO

Colorectal cancer (CRC) is the third most diagnosed cancer and the second deadliest cancer worldwide representing a major public health problem. In recent years, increasing evidence has shown that microRNA (miRNA) can control the expression of targeted human messenger RNA (mRNA) by reducing their abundance or translation, acting as oncogenes or tumor suppressors in various cancers, including CRC. Due to the significant up-regulation of oncogenic miRNAs in CRC, elucidating the underlying mechanism and identifying dysregulated miRNA targets may provide a basis for improving current therapeutic interventions. In this paper, we proposed Gra-CRC-miRTar, a pre-trained nucleotide-to-graph neural network framework, for identifying potential miRNA targets in CRC. Different from previous studies, we constructed two pre-trained models to encode RNA sequences and transformed them into de Bruijn graphs. We employed different graph neural networks to learn the latent representations. The embeddings generated from de Bruijn graphs were then fed into a Multilayer Perceptron (MLP) to perform the prediction tasks. Our extensive experiments show that Gra-CRC-miRTar achieves better performance than other deep learning algorithms and existing predictors. In addition, our analyses also successfully revealed 172 out of 201 functional interactions through experimentally validated miRNA-mRNA pairs in CRC. Collectively, our effort provides an accurate and efficient framework to identify potential miRNA targets in CRC, which can also be used to reveal miRNA target interactions in other malignancies, facilitating the development of novel therapeutics.

9.
Int ACM SIGIR Conf Res Dev Inf Retr ; 2023: 2501-2505, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38352126

RESUMO

Scientific document classification is a critical task for a wide range of applications, but the cost of collecting human-labeled data can be prohibitive. We study scientific document classification using label names only. In scientific domains, label names often include domain-specific concepts that may not appear in the document corpus, making it difficult to match labels and documents precisely. To tackle this issue, we propose WanDeR, which leverages dense retrieval to perform matching in the embedding space to capture the semantics of label names. We further design the label name expansion module to enrich its representations. Lastly, a self-training step is used to refine the predictions. The experiments on three datasets show that WanDeR outperforms the best baseline by 11.9%. Our code will be published at https://github.com/ritaranx/wander.

10.
AMIA Jt Summits Transl Sci Proc ; 2023: 582-591, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37350881

RESUMO

Electronic health records (EHR) data contain rich information about patients' health conditions including diagnosis, procedures, medications and etc., which have been widely used to facilitate digital medicine. Despite its importance, it is often non-trivial to learn useful representations for patients' visits that support downstream clinical predictions, as each visit contains massive and diverse medical codes. As a result, the complex interactions among medical codes are often not captured, which leads to substandard predictions. To better model these complex relations, we leverage hypergraphs, which go beyond pairwise relations to jointly learn the representations for visits and medical codes. We also propose to use the self-attention mechanism to automatically identify the most relevant medical codes for each visit based on the downstream clinical predictions with better generalization power. Experiments on two EHR datasets show that our proposed method not only yields superior performance, but also provides reasonable insights towards the target tasks.

11.
Adv Neural Inf Process Syst ; 36: 13308-13325, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39130614

RESUMO

Graphs are widely used to model interconnected entities and improve downstream predictions in various real-world applications. However, real-world graphs nowadays are often associated with complex attributes on multiple types of nodes and even links that are hard to model uniformly, while the widely used graph neural networks (GNNs) often require sufficient training toward specific downstream predictions to achieve strong performance. In this work, we take a fundamentally different approach than GNNs, to simultaneously achieve deep joint modeling of complex attributes and flexible structures of real-world graphs and obtain unsupervised generic graph representations that are not limited to specific downstream predictions. Our framework, built on a natural integration of language models (LMs) and random walks (RWs), is straightforward, powerful and data-efficient. Specifically, we first perform attributed RWs on the graph and design an automated program to compose roughly meaningful textual sequences directly from the attributed RWs; then we fine-tune an LM using the RW-based textual sequences and extract embedding vectors from the LM, which encapsulates both attribute semantics and graph structures. In our experiments, we evaluate the learned node embeddings towards different downstream prediction tasks on multiple real-world attributed graph datasets and observe significant improvements over a comprehensive set of state-of-the-art unsupervised node embedding methods. We believe this work opens a door for more sophisticated technical designs and empirical evaluations toward the leverage of LMs for the modeling of real-world graphs.

12.
IEEE Int Conf Healthc Inform ; 2023: 128-137, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38332952

RESUMO

The utilization of web search activity for pandemic forecasting has significant implications for managing disease spread and informing policy decisions. However, web search records tend to be noisy and influenced by geographical location, making it difficult to develop large-scale models. While regularized linear models have been effective in predicting the spread of respiratory illnesses like COVID-19, they are limited to specific locations. The lack of incorporation of neighboring areas' data and the inability to transfer models to new locations with limited data has impeded further progress. To address these limitations, this study proposes a novel self-supervised message-passing neural network (SMPNN) framework for modeling local and cross-location dynamics in pandemic forecasting. The SMPNN framework utilizes an MPNN module to learn cross-location dependencies through self-supervised learning and improve local predictions with graph-generated features. The framework is designed as an end-to-end solution and is compared with state-of-the-art statistical and deep learning models using COVID-19 data from England and the US. The results of the study demonstrate that the SMPNN model outperforms other models by achieving up to a 6.9% improvement in prediction accuracy and lower prediction errors during the early stages of disease outbreaks. This approach represents a significant advancement in disease surveillance and forecasting, providing a novel methodology, datasets, and insights that combine web search data and spatial information. The proposed SMPNN framework offers a promising avenue for modeling the spread of pandemics, leveraging both local and cross-location information, and has the potential to inform public health policy decisions.

13.
Int ACM SIGIR Conf Res Dev Inf Retr ; 2023: 2052-2056, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38352127

RESUMO

Medical decision-making processes can be enhanced by comprehensive biomedical knowledge bases, which require fusing knowledge graphs constructed from different sources via a uniform index system. The index system often organizes biomedical terms in a hierarchy to provide the aligned entities with fine-grained granularity. To address the challenge of scarce supervision in the biomedical knowledge fusion (BKF) task, researchers have proposed various unsupervised methods. However, these methods heavily rely on ad-hoc lexical and structural matching algorithms, which fail to capture the rich semantics conveyed by biomedical entities and terms. Recently, neural embedding models have proved effective in semantic-rich tasks, but they rely on sufficient labeled data to be adequately trained. To bridge the gap between the scarce-labeled BKF and neural embedding models, we propose HiPrompt, a supervision-efficient knowledge fusion framework that elicits the few-shot reasoning ability of large language models through hierarchy-oriented prompts. Empirical results on the collected KG-Hi-BKF benchmark datasets demonstrate the effectiveness of HiPrompt.

14.
Circ Cardiovasc Imaging ; 16(12): e014533, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38073535

RESUMO

In addition to the traditional clinical risk factors, an increasing amount of imaging biomarkers have shown value for cardiovascular risk prediction. Clinical and imaging data are captured from a variety of data sources during multiple patient encounters and are often analyzed independently. Initial studies showed that fusion of both clinical and imaging features results in superior prognostic performance compared with traditional scores. There are different approaches to fusion modeling, combining multiple data resources to optimize predictions, each with its own advantages and disadvantages. However, manual extraction of clinical and imaging data is time and labor intensive and often not feasible in clinical practice. An automated approach for clinical and imaging data extraction is highly desirable. Convolutional neural networks and natural language processing can be utilized for the extraction of electronic medical record data, imaging studies, and free-text data. This review outlines the current status of cardiovascular risk prediction and fusion modeling; and in addition gives an overview of different artificial intelligence approaches to automatically extract data from images and electronic medical records for this purpose.


Assuntos
Inteligência Artificial , Redes Neurais de Computação , Humanos , Registros Eletrônicos de Saúde , Processamento de Linguagem Natural , Diagnóstico por Imagem
15.
Adv Neural Inf Process Syst ; 36: 56946-56978, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39144377

RESUMO

Pre-training on graph neural networks (GNNs) aims to learn transferable knowledge for downstream tasks with unlabeled data, and it has recently become an active research area. The success of graph pre-training models is often attributed to the massive amount of input data. In this paper, however, we identify the curse of big data phenomenon in graph pre-training: more training data do not necessarily lead to better downstream performance. Motivated by this observation, we propose a better-with-less framework for graph pre-training: fewer, but carefully chosen data are fed into a GNN model to enhance pre-training. The proposed pre-training pipeline is called the data-active graph pre-training (APT) framework, and is composed of a graph selector and a pre-training model. The graph selector chooses the most representative and instructive data points based on the inherent properties of graphs as well as predictive uncertainty. The proposed predictive uncertainty, as feedback from the pre-training model, measures the confidence level of the model in the data. When fed with the chosen data, on the other hand, the pre-training model grasps an initial understanding of the new, unseen data, and at the same time attempts to remember the knowledge learned from previous data. Therefore, the integration and interaction between these two components form a unified framework (APT), in which graph pre-training is performed in a progressive and iterative way. Experiment results show that the proposed APT is able to obtain an efficient pre-training model with fewer training data and better downstream performance.

16.
Proc AAAI Conf Artif Intell ; 37(9): 10611-10619, 2023 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38333625

RESUMO

Training deep neural networks (DNNs) with limited supervision has been a popular research topic as it can significantly alleviate the annotation burden. Self-training has been successfully applied in semi-supervised learning tasks, but one drawback of self-training is that it is vulnerable to the label noise from incorrect pseudo labels. Inspired by the fact that samples with similar labels tend to share similar representations, we develop a neighborhood-based sample selection approach to tackle the issue of noisy pseudo labels. We further stabilize self-training via aggregating the predictions from different rounds during sample selection. Experiments on eight tasks show that our proposed method outperforms the strongest self-training baseline with 1.83% and 2.51% performance gain for text and graph datasets on average. Our further analysis demonstrates that our proposed data selection strategy reduces the noise of pseudo labels by 36.8% and saves 57.3% of the time when compared with the best baseline. Our code and appendices will be uploaded to https://github.com/ritaranx/NeST.

17.
KDD ; 2023: 142-153, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38333106

RESUMO

In recent years, graph pre-training has gained significant attention, focusing on acquiring transferable knowledge from unlabeled graph data to improve downstream performance. Despite these recent endeavors, the problem of negative transfer remains a major concern when utilizing graph pre-trained models to downstream tasks. Previous studies made great efforts on the issue of what to pre-train and how to pre-train by designing a variety of graph pre-training and fine-tuning strategies. However, there are cases where even the most advanced "pre-train and fine-tune" paradigms fail to yield distinct benefits. This paper introduces a generic framework W2PGNN to answer the crucial question of when to pre-train (i.e., in what situations could we take advantage of graph pre-training) before performing effortful pre-training or fine-tuning. We start from a new perspective to explore the complex generative mechanisms from the pre-training data to downstream data. In particular, W2PGNN first fits the pre-training data into graphon bases, each element of graphon basis (i.e., a graphon) identifies a fundamental transferable pattern shared by a collection of pre-training graphs. All convex combinations of graphon bases give rise to a generator space, from which graphs generated form the solution space for those downstream data that can benefit from pre-training. In this manner, the feasibility of pre-training can be quantified as the generation probability of the downstream data from any generator in the generator space. W2PGNN offers three broad applications: providing the application scope of graph pre-trained models, quantifying the feasibility of pre-training, and assistance in selecting pre-training data to enhance downstream performance. We provide a theoretically sound solution for the first application and extensive empirical justifications for the latter two applications.

18.
IEEE Trans Med Imaging ; 42(2): 493-506, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36318557

RESUMO

Mapping the connectome of the human brain using structural or functional connectivity has become one of the most pervasive paradigms for neuroimaging analysis. Recently, Graph Neural Networks (GNNs) motivated from geometric deep learning have attracted broad interest due to their established power for modeling complex networked data. Despite their superior performance in many fields, there has not yet been a systematic study of how to design effective GNNs for brain network analysis. To bridge this gap, we present BrainGB, a benchmark for brain network analysis with GNNs. BrainGB standardizes the process by (1) summarizing brain network construction pipelines for both functional and structural neuroimaging modalities and (2) modularizing the implementation of GNN designs. We conduct extensive experiments on datasets across cohorts and modalities and recommend a set of general recipes for effective GNN designs on brain networks. To support open and reproducible research on GNN-based brain network analysis, we host the BrainGB website at https://braingb.us with models, tutorials, examples, as well as an out-of-box Python package. We hope that this work will provide useful empirical evidence and offer insights for future research in this novel and promising direction.


Assuntos
Benchmarking , Conectoma , Humanos , Encéfalo/diagnóstico por imagem , Redes Neurais de Computação , Neuroimagem
19.
ArXiv ; 2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37332568

RESUMO

Biological networks are commonly used in biomedical and healthcare domains to effectively model the structure of complex biological systems with interactions linking biological entities. However, due to their characteristics of high dimensionality and low sample size, directly applying deep learning models on biological networks usually faces severe overfitting. In this work, we propose R-MIXUP, a Mixup-based data augmentation technique that suits the symmetric positive definite (SPD) property of adjacency matrices from biological networks with optimized training efficiency. The interpolation process in R-MIXUP leverages the log-Euclidean distance metrics from the Riemannian manifold, effectively addressing the swelling effect and arbitrarily incorrect label issues of vanilla Mixup. We demonstrate the effectiveness of R-MIXUP with five real-world biological network datasets on both regression and classification tasks. Besides, we derive a commonly ignored necessary condition for identifying the SPD matrices of biological networks and empirically study its influence on the model performance. The code implementation can be found in Appendix E.

20.
Adv Neural Inf Process Syst ; 36: 23499-23519, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39130613

RESUMO

Images contain rich relational knowledge that can help machines understand the world. Existing methods on visual knowledge extraction often rely on the pre-defined format (e.g., sub-verb-obj tuples) or vocabulary (e.g., relation types), restricting the expressiveness of the extracted knowledge. In this work, we take a first exploration to a new paradigm of open visual knowledge extraction. To achieve this, we present OpenVik which consists of an open relational region detector to detect regions potentially containing relational knowledge and a visual knowledge generator that generates format-free knowledge by prompting the large multimodality model with the detected region of interest. We also explore two data enhancement techniques for diversifying the generated format-free visual knowledge. Extensive knowledge quality evaluations highlight the correctness and uniqueness of the extracted open visual knowledge by OpenVik. Moreover, integrating our extracted knowledge across various visual reasoning applications shows consistent improvements, indicating the real-world applicability of OpenVik.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA