Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.719
Filtrar
Más filtros

Tipo del documento
Publication year range
1.
Brief Bioinform ; 25(5)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39101499

RESUMEN

Genome-scale metabolic models (GEMs) are powerful tools for predicting cellular metabolic and physiological states. However, there are still missing reactions in GEMs due to incomplete knowledge. Recent gaps filling methods suggest directly predicting missing responses without relying on phenotypic data. However, they do not differentiate between substrates and products when constructing the prediction models, which affects the predictive performance of the models. In this paper, we propose a hyperedge prediction model that distinguishes substrates and products based on dual-scale fused hypergraph convolution, DSHCNet, for inferring the missing reactions to effectively fill gaps in the GEM. First, we model each hyperedge as a heterogeneous complete graph and then decompose it into three subgraphs at both homogeneous and heterogeneous scales. Then we design two graph convolution-based models to, respectively, extract features of the vertices in two scales, which are then fused via the attention mechanism. Finally, the features of all vertices are further pooled to generate the representative feature of the hyperedge. The strategy of graph decomposition in DSHCNet enables the vertices to engage in message passing independently at both scales, thereby enhancing the capability of information propagation and making the obtained product and substrate features more distinguishable. The experimental results show that the average recovery rate of missing reactions obtained by DSHCNet is at least 11.7% higher than that of the state-of-the-art methods, and that the gap-filled GEMs based on our DSHCNet model achieve the best prediction performance, demonstrating the superiority of our method.


Asunto(s)
Redes y Vías Metabólicas , Algoritmos , Modelos Biológicos , Genoma , Biología Computacional/métodos
2.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38426320

RESUMEN

Protein subcellular localization (PSL) is very important in order to understand its functions, and its movement between subcellular niches within cells plays fundamental roles in biological process regulation. Mass spectrometry-based spatio-temporal proteomics technologies can help provide new insights of protein translocation, but bring the challenge in identifying reliable protein translocation events due to the noise interference and insufficient data mining. We propose a semi-supervised graph convolution network (GCN)-based framework termed TransGCN that infers protein translocation events from spatio-temporal proteomics. Based on expanded multiple distance features and joint graph representations of proteins, TransGCN utilizes the semi-supervised GCN to enable effective knowledge transfer from proteins with known PSLs for predicting protein localization and translocation. Our results demonstrate that TransGCN outperforms current state-of-the-art methods in identifying protein translocations, especially in coping with batch effects. It also exhibited excellent predictive accuracy in PSL prediction. TransGCN is freely available on GitHub at https://github.com/XuejiangGuo/TransGCN.


Asunto(s)
Habilidades de Afrontamiento , Proteómica , Minería de Datos , Espectrometría de Masas , Transporte de Proteínas
3.
Proc Natl Acad Sci U S A ; 120(25): e2304589120, 2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37307444

RESUMEN

We introduce a framework to study discrete-variable (DV) quantum systems based on qudits. It relies on notions of a mean state (MS), a minimal stabilizer-projection state (MSPS), and a new convolution. Some interesting consequences are: The MS is the closest MSPS to a given state with respect to the relative entropy; the MS is extremal with respect to the von Neumann entropy, demonstrating a "maximal entropy principle in DV systems." We obtain a series of inequalities for quantum entropies and for Fisher information based on convolution, giving a "second law of thermodynamics for quantum convolutions." We show that the convolution of two stabilizer states is a stabilizer state. We establish a central limit theorem, based on iterating the convolution of a zero-mean quantum state, and show this converges to its MS. The rate of convergence is characterized by the "magic gap," which we define in terms of the support of the characteristic function of the state. We elaborate on two examples: the DV beam splitter and the DV amplifier.

4.
Brief Bioinform ; 24(4)2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37385595

RESUMEN

Allergies have become an emerging public health problem worldwide. The most effective way to prevent allergies is to find the causative allergen at the source and avoid re-exposure. However, most of the current computational methods used to identify allergens were based on homology or conventional machine learning methods, which were inefficient and still had room to be improved for the detection of allergens with low homology. In addition, few methods based on deep learning were reported, although deep learning has been successfully applied to several tasks in protein sequence analysis. In the present work, a deep neural network-based model, called DeepAlgPro, was proposed to identify allergens. We showed its great accuracy and applicability to large-scale forecasts by comparing it to other available tools. Additionally, we used ablation experiments to demonstrate the critical importance of the convolutional module in our model. Moreover, further analyses showed that epitope features contributed to model decision-making, thus improving the model's interpretability. Finally, we found that DeepAlgPro was capable of detecting potential new allergens. Overall, DeepAlgPro can serve as powerful software for identifying allergens.


Asunto(s)
Aprendizaje Profundo , Hipersensibilidad , Humanos , Alérgenos , Redes Neurales de la Computación , Proteínas/metabolismo
5.
Brief Bioinform ; 24(6)2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37779247

RESUMEN

Accurate identification of protein-protein interaction (PPI) sites remains a computational challenge. We propose Spatom, a novel framework for PPI site prediction. This framework first defines a weighted digraph for a protein structure to precisely characterize the spatial contacts of residues, then performs a weighted digraph convolution to aggregate both spatial local and global information and finally adds an improved graph attention layer to drive the predicted sites to form more continuous region(s). Spatom was tested on a diverse set of challenging protein-protein complexes and demonstrated the best performance among all the compared methods. Furthermore, when tested on multiple popular proteins in a case study, Spatom clearly identifies the interaction interfaces and captures the majority of hotspots. Spatom is expected to contribute to the understanding of protein interactions and drug designs targeting protein binding.


Asunto(s)
Redes Neurales de la Computación , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Unión Proteica , Proteínas/metabolismo
6.
Brief Bioinform ; 24(5)2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37544658

RESUMEN

MOTIVATION: Recent advances in spatially resolved transcriptomics (ST) technologies enable the measurement of gene expression profiles while preserving cellular spatial context. Linking gene expression of cells with their spatial distribution is essential for better understanding of tissue microenvironment and biological progress. However, effectively combining gene expression data with spatial information to identify spatial domains remains challenging. RESULTS: To deal with the above issue, in this paper, we propose a novel unsupervised learning framework named STMGCN for identifying spatial domains using multi-view graph convolution networks (MGCNs). Specifically, to fully exploit spatial information, we first construct multiple neighbor graphs (views) with different similarity measures based on the spatial coordinates. Then, STMGCN learns multiple view-specific embeddings by combining gene expressions with each neighbor graph through graph convolution networks. Finally, to capture the importance of different graphs, we further introduce an attention mechanism to adaptively fuse view-specific embeddings and thus derive the final spot embedding. STMGCN allows for the effective utilization of spatial context to enhance the expressive power of the latent embeddings with multiple graph convolutions. We apply STMGCN on two simulation datasets and five real spatial transcriptomics datasets with different resolutions across distinct platforms. The experimental results demonstrate that STMGCN obtains competitive results in spatial domain identification compared with five state-of-the-art methods, including spatial and non-spatial alternatives. Besides, STMGCN can detect spatially variable genes with enriched expression patterns in the identified domains. Overall, STMGCN is a powerful and efficient computational framework for identifying spatial domains in spatial transcriptomics data.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Simulación por Computador
7.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36526276

RESUMEN

Increasing studies have proved that microRNAs (miRNAs) are critical biomarkers in the development of human complex diseases. Identifying disease-related miRNAs is beneficial to disease prevention, diagnosis and remedy. Based on the assumption that similar miRNAs tend to associate with similar diseases, various computational methods have been developed to predict novel miRNA-disease associations (MDAs). However, selecting proper features for similarity calculation is a challenging task because of data deficiencies in biomedical science. In this study, we propose a deep learning-based computational method named MAGCN to predict potential MDAs without using any similarity measurements. Our method predicts novel MDAs based on known lncRNA-miRNA interactions via graph convolution networks with multichannel attention mechanism and convolutional neural network combiner. Extensive experiments show that the average area under the receiver operating characteristic values obtained by our method under 2-fold, 5-fold and 10-fold cross-validations are 0.8994, 0.9032 and 0.9044, respectively. When compared with five state-of-the-art methods, MAGCN shows improvement in terms of prediction accuracy. In addition, we conduct case studies on three diseases to discover their related miRNAs, and find that all the top 50 predictions for all the three diseases have been supported by established databases. The comprehensive results demonstrate that our method is a reliable tool in detecting new disease-related miRNAs.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , Algoritmos , Biología Computacional/métodos , Bases de Datos Genéticas , MicroARNs/genética , ARN Largo no Codificante/genética , Aprendizaje Profundo
8.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36526282

RESUMEN

Identifying unknown protein functional modules, such as protein complexes and biological pathways, from protein-protein interaction (PPI) networks, provides biologists with an opportunity to efficiently understand cellular function and organization. Finding complex nonlinear relationships in underlying functional modules may involve a long-chain of PPI and pose great challenges in a PPI network with an unevenly sparse and dense node distribution. To overcome these challenges, we propose AdaPPI, an adaptive convolution graph network in PPI networks to predict protein functional modules. We first suggest an attributed graph node presentation algorithm. It can effectively integrate protein gene ontology attributes and network topology, and adaptively aggregates low- or high-order graph structural information according to the node distribution by considering graph node smoothness. Based on the obtained node representations, core cliques and expansion algorithms are applied to find functional modules in PPI networks. Comprehensive performance evaluations and case studies indicate that the framework significantly outperforms state-of-the-art methods. We also presented potential functional modules based on their confidence.


Asunto(s)
Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Algoritmos , Proteínas/genética , Proteínas/metabolismo
9.
Brief Bioinform ; 25(1)2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-38243694

RESUMEN

The correct prediction of disease-associated miRNAs plays an essential role in disease prevention and treatment. Current computational methods to predict disease-associated miRNAs construct different miRNA views and disease views based on various miRNA properties and disease properties and then integrate the multiviews to predict the relationship between miRNAs and diseases. However, most existing methods ignore the information interaction among the views and the consistency of miRNA features (disease features) across multiple views. This study proposes a computational method based on multiple hypergraph contrastive learning (MHCLMDA) to predict miRNA-disease associations. MHCLMDA first constructs multiple miRNA hypergraphs and disease hypergraphs based on various miRNA similarities and disease similarities and performs hypergraph convolution on each hypergraph to capture higher order interactions between nodes, followed by hypergraph contrastive learning to learn the consistent miRNA feature representation and disease feature representation under different views. Then, a variational auto-encoder is employed to extract the miRNA and disease features in known miRNA-disease association relationships. Finally, MHCLMDA fuses the miRNA and disease features from different views to predict miRNA-disease associations. The parameters of the model are optimized in an end-to-end way. We applied MHCLMDA to the prediction of human miRNA-disease association. The experimental results show that our method performs better than several other state-of-the-art methods in terms of the area under the receiver operating characteristic curve and the area under the precision-recall curve.


Asunto(s)
MicroARNs , Humanos , MicroARNs/genética , Algoritmos , Biología Computacional/métodos , Curva ROC
10.
Methods ; 229: 71-81, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38909974

RESUMEN

Identifying miRNA-disease associations (MDAs) is crucial for improving the diagnosis and treatment of various diseases. However, biological experiments can be time-consuming and expensive. To overcome these challenges, computational approaches have been developed, with Graph Convolutional Network (GCN) showing promising results in MDA prediction. The success of GCN-based methods relies on learning a meaningful spatial operator to extract effective node feature representations. To enhance the inference of MDAs, we propose a novel method called PGCNMDA, which employs graph convolutional networks with a learning graph spatial operator from paths. This approach enables the generation of meaningful spatial convolutions from paths in GCN, leading to improved prediction performance. On HMDD v2.0, PGCNMDA obtains a mean AUC of 0.9229 and an AUPRC of 0.9206 under 5-fold cross-validation (5-CV), and a mean AUC of 0.9235 and an AUPRC of 0.9212 under 10-fold cross-validation (10-CV), respectively. Additionally, the AUC of PGCNMDA also reaches 0.9238 under global leave-one-out cross-validation (GLOOCV). On HMDD v3.2, PGCNMDA obtains a mean AUC of 0.9413 and an AUPRC of 0.9417 under 5-CV, and a mean AUC of 0.9419 and an AUPRC of 0.9425 under 10-CV, respectively. Furthermore, the AUC of PGCNMDA also reaches 0.9415 under GLOOCV. The results show that PGCNMDA is superior to other compared methods. In addition, the case studies on pancreatic neoplasms, thyroid neoplasms and leukemia show that 50, 50 and 48 of the top 50 predicted miRNAs linked to these diseases are confirmed, respectively. It further validates the effectiveness and feasibility of PGCNMDA in practical applications.


Asunto(s)
MicroARNs , Humanos , MicroARNs/genética , Biología Computacional/métodos , Redes Neurales de la Computación , Predisposición Genética a la Enfermedad , Área Bajo la Curva , Neoplasias Pancreáticas/genética , Algoritmos
11.
Cereb Cortex ; 34(3)2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38494887

RESUMEN

The early diagnosis of autism spectrum disorder (ASD) has been extensively facilitated through the utilization of resting-state fMRI (rs-fMRI). With rs-fMRI, the functional brain network (FBN) has gained much attention in diagnosing ASD. As a promising strategy, graph convolutional networks (GCN) provide an attractive approach to simultaneously extract FBN features and facilitate ASD identification, thus replacing the manual feature extraction from FBN. Previous GCN studies primarily emphasized the exploration of topological simultaneously connection weights of the estimated FBNs while only focusing on the single connection pattern. However, this approach fails to exploit the potential complementary information offered by different connection patterns of FBNs, thereby inherently limiting the performance. To enhance the diagnostic performance, we propose a multipattern graph convolution network (MPGCN) that integrates multiple connection patterns to improve the accuracy of ASD diagnosis. As an initial endeavor, we endeavored to integrate information from multiple connection patterns by incorporating multiple graph convolution modules. The effectiveness of the MPGCN approach is evaluated by analyzing rs-fMRI scans from a cohort of 92 subjects sourced from the publicly accessible Autism Brain Imaging Data Exchange database. Notably, the experiment demonstrates that our model achieves an accuracy of 91.1% and an area under ROC curve score of 0.9742. The implementation codes are available at https://github.com/immutableJackz/MPGCN.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Humanos , Trastorno del Espectro Autista/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Bases de Datos Factuales , Curva ROC
12.
Proc Natl Acad Sci U S A ; 119(40): e2201854119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161906

RESUMEN

Exploiting data invariances is crucial for efficient learning in both artificial and biological neural circuits. Understanding how neural networks can discover appropriate representations capable of harnessing the underlying symmetries of their inputs is thus crucial in machine learning and neuroscience. Convolutional neural networks, for example, were designed to exploit translation symmetry, and their capabilities triggered the first wave of deep learning successes. However, learning convolutions directly from translation-invariant data with a fully connected network has so far proven elusive. Here we show how initially fully connected neural networks solving a discrimination task can learn a convolutional structure directly from their inputs, resulting in localized, space-tiling receptive fields. These receptive fields match the filters of a convolutional network trained on the same task. By carefully designing data models for the visual scene, we show that the emergence of this pattern is triggered by the non-Gaussian, higher-order local structure of the inputs, which has long been recognized as the hallmark of natural images. We provide an analytical and numerical characterization of the pattern formation mechanism responsible for this phenomenon in a simple model and find an unexpected link between receptive field formation and tensor decomposition of higher-order input correlations. These results provide a perspective on the development of low-level feature detectors in various sensory modalities and pave the way for studying the impact of higher-order statistics on learning in neural networks.


Asunto(s)
Aprendizaje Automático , Redes Neurales de la Computación , Neurociencias
13.
Nano Lett ; 24(8): 2589-2595, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38252875

RESUMEN

Surface topography, or height profile, is a critical property for various micro- and nanostructured materials and devices, as well as biological systems. At the nanoscale, atomic force microscopy (AFM) is the tool of choice for surface profiling due to its capability to noninvasively map the topography of almost all types of samples. However, this method suffers from one drawback: the convolution of the nanoprobe's shape in the height profile of the samples, which is especially severe for sharp protrusion features. Here, we report a deep learning (DL) approach to overcome this limit. Adopting an image-to-image translation methodology, we use data sets of tip-convoluted and deconvoluted image pairs to train an encoder-decoder based deep convolutional neural network. The trained network successfully removes the tip convolution from AFM topographic images of various nanocorrugated surfaces and recovers the true, precise 3D height profiles of these samples.

14.
Brief Bioinform ; 23(3)2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35323894

RESUMEN

While the technologies of ribonucleic acid-sequence (RNA-seq) and transcript assembly analysis have continued to improve, a novel topology of RNA transcript was uncovered in the last decade and is called circular RNA (circRNA). Recently, researchers have revealed that they compete with messenger RNA (mRNA) and long noncoding for combining with microRNA in gene regulation. Therefore, circRNA was assumed to be associated with complex disease and discovering the relationship between them would contribute to medical research. However, the work of identifying the association between circRNA and disease in vitro takes a long time and usually without direction. During these years, more and more associations were verified by experiments. Hence, we proposed a computational method named identifying circRNA-disease association based on graph representation learning (iGRLCDA) for the prediction of the potential association of circRNA and disease, which utilized a deep learning model of graph convolution network (GCN) and graph factorization (GF). In detail, iGRLCDA first derived the hidden feature of known associations between circRNA and disease using the Gaussian interaction profile (GIP) kernel combined with disease semantic information to form a numeric descriptor. After that, it further used the deep learning model of GCN and GF to extract hidden features from the descriptor. Finally, the random forest classifier is introduced to identify the potential circRNA-disease association. The five-fold cross-validation of iGRLCDA shows strong competitiveness in comparison with other excellent prediction models at the gold standard data and achieved an average area under the receiver operating characteristic curve of 0.9289 and an area under the precision-recall curve of 0.9377. On reviewing the prediction results from the relevant literature, 22 of the top 30 predicted circRNA-disease associations were noted in recent published papers. These exceptional results make us believe that iGRLCDA can provide reliable circRNA-disease associations for medical research and reduce the blindness of wet-lab experiments.


Asunto(s)
MicroARNs , ARN Circular , Algoritmos , Biología Computacional/métodos , MicroARNs/genética , Curva ROC
15.
Brief Bioinform ; 23(6)2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36209437

RESUMEN

Long non-coding RNA (lncRNA) plays important roles in a series of biological processes. The transcription of lncRNA is regulated by its promoter. Hence, accurate identification of lncRNA promoter will be helpful to understand its regulatory mechanisms. Since experimental techniques remain time consuming for gnome-wide promoter identification, developing computational tools to identify promoters are necessary. However, only few computational methods have been proposed for lncRNA promoter prediction and their performances still have room to be improved. In the present work, a convolutional neural network based model, called DeepLncPro, was proposed to identify lncRNA promoters in human and mouse. Comparative results demonstrated that DeepLncPro was superior to both state-of-the-art machine learning methods and existing models for identifying lncRNA promoters. Furthermore, DeepLncPro has the ability to extract and analyze transcription factor binding motifs from lncRNAs, which made it become an interpretable model. These results indicate that the DeepLncPro can server as a powerful tool for identifying lncRNA promoters. An open-source tool for DeepLncPro was provided at https://github.com/zhangtian-yang/DeepLncPro.


Asunto(s)
ARN Largo no Codificante , Humanos , Animales , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Biología Computacional/métodos , Redes Neurales de la Computación , Regiones Promotoras Genéticas , Algoritmos
16.
Brief Bioinform ; 23(5)2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-35907779

RESUMEN

Circular RNA (circRNA) is closely involved in physiological and pathological processes of many diseases. Discovering the associations between circRNAs and diseases is of great significance. Due to the high-cost to verify the circRNA-disease associations by wet-lab experiments, computational approaches for predicting the associations become a promising research direction. In this paper, we propose a method, MDGF-MCEC, based on multi-view dual attention graph convolution network (GCN) with cooperative ensemble learning to predict circRNA-disease associations. First, MDGF-MCEC constructs two disease relation graphs and two circRNA relation graphs based on different similarities. Then, the relation graphs are fed into a multi-view GCN for representation learning. In order to learn high discriminative features, a dual-attention mechanism is introduced to adjust the contribution weights, at both channel level and spatial level, of different features. Based on the learned embedding features of diseases and circRNAs, nine different feature combinations between diseases and circRNAs are treated as new multi-view data. Finally, we construct a multi-view cooperative ensemble classifier to predict the associations between circRNAs and diseases. Experiments conducted on the CircR2Disease database demonstrate that the proposed MDGF-MCEC model achieves a high area under curve of 0.9744 and outperforms the state-of-the-art methods. Promising results are also obtained from experiments on the circ2Disease and circRNADisease databases. Furthermore, the predicted associated circRNAs for hepatocellular carcinoma and gastric cancer are supported by the literature. The code and dataset of this study are available at https://github.com/ABard0/MDGF-MCEC.


Asunto(s)
ARN Circular , Neoplasias Gástricas , Humanos , Péptidos y Proteínas de Señalización Intercelular , Aprendizaje Automático , Neoplasias Gástricas/genética
17.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34849567

RESUMEN

MOTIVATION: Understanding chemical-gene interactions (CGIs) is crucial for screening drugs. Wet experiments are usually costly and laborious, which limits relevant studies to a small scale. On the contrary, computational studies enable efficient in-silico exploration. For the CGI prediction problem, a common method is to perform systematic analyses on a heterogeneous network involving various biomedical entities. Recently, graph neural networks become popular in the field of relation prediction. However, the inherent heterogeneous complexity of biological interaction networks and the massive amount of data pose enormous challenges. This paper aims to develop a data-driven model that is capable of learning latent information from the interaction network and making correct predictions. RESULTS: We developed BioNet, a deep biological networkmodel with a graph encoder-decoder architecture. The graph encoder utilizes graph convolution to learn latent information embedded in complex interactions among chemicals, genes, diseases and biological pathways. The learning process is featured by two consecutive steps. Then, embedded information learnt by the encoder is then employed to make multi-type interaction predictions between chemicals and genes with a tensor decomposition decoder based on the RESCAL algorithm. BioNet includes 79 325 entities as nodes, and 34 005 501 relations as edges. To train such a massive deep graph model, BioNet introduces a parallel training algorithm utilizing multiple Graphics Processing Unit (GPUs). The evaluation experiments indicated that BioNet exhibits outstanding prediction performance with a best area under Receiver Operating Characteristic (ROC) curve of 0.952, which significantly surpasses state-of-theart methods. For further validation, top predicted CGIs of cancer and COVID-19 by BioNet were verified by external curated data and published literature.


Asunto(s)
Biología Computacional , Simulación por Computador , Modelos Biológicos , Redes Neurales de la Computación
18.
Anal Biochem ; 690: 115491, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38460901

RESUMEN

Bioactive peptides can hinder oxidative processes and microbial spoilage in foodstuffs and play important roles in treating diverse diseases and disorders. While most of the methods focus on single-functional bioactive peptides and have obtained promising prediction performance, it is still a significant challenge to accurately detect complex and diverse functions simultaneously with the quick increase of multi-functional bioactive peptides. In contrast to previous research on multi-functional bioactive peptide prediction based solely on sequence, we propose a novel multimodal dual-branch (MMDB) lightweight deep learning model that designs two different branches to effectively capture the complementary information of peptide sequence and structural properties. Specifically, a multi-scale dilated convolution with Bi-LSTM branch is presented to effectively model the different scales sequence properties of peptides while a multi-layer convolution branch is proposed to capture structural information. To the best of our knowledge, this is the first effective extraction of peptide sequence features using multi-scale dilated convolution without parameter increase. Multimodal features from both branches are integrated via a fully connected layer for multi-label classification. Compared to state-of-the-art methods, our MMDB model exhibits competitive results across metrics, with a 9.1% Coverage increase and 5.3% and 3.5% improvements in Precision and Accuracy, respectively.

19.
J Comput Neurosci ; 52(1): 39-71, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38381252

RESUMEN

The computational resources of a neuromorphic network model introduced earlier are investigated in the context of such hierarchical systems as the mammalian visual cortex. It is argued that a form of ubiquitous spontaneous local convolution, driven by spontaneously arising wave-like activity-which itself promotes local Hebbian modulation-enables logical gate-like neural motifs to form into hierarchical feed-forward structures of the Hubel-Wiesel type. Extra-synaptic effects are shown to play a significant rôle in these processes. The type of logic that emerges is not Boolean, confirming and extending earlier findings on the logic of schizophrenia.


Asunto(s)
Modelos Neurológicos , Corteza Visual , Animales , Red Nerviosa , Mamíferos
20.
J Comput Neurosci ; 52(3): 223-243, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39083150

RESUMEN

The computational resources of a neuromorphic network model introduced earlier were investigated in the first paper of this series. It was argued that a form of ubiquitous spontaneous local convolution enabled logical gate-like neural motifs to form into hierarchical feed-forward structures of the Hubel-Wiesel type. Here we investigate concomitant data-like structures and their dynamic rôle in memory formation, retrieval, and replay. The mechanisms give rise to the need for general inhibitory sculpting, and the simulation of the replay of episodic memories, well known in humans and recently observed in rats. Other consequences include explanations of such findings as the directional flows of neural waves in memory formation and retrieval, visual anomalies and memory deficits in schizophrenia, and the operation of GABA agonist drugs in suppressing episodic memories. We put forward the hypothesis that all neural logical operations and feature extractions are of the convolutional hierarchical type described here and in the earlier paper, and exemplified by the Hubel-Wiesel model of the visual cortex, but that in more general cases the precise geometric layering might be obscured and so far undetected.


Asunto(s)
Memoria Episódica , Modelos Neurológicos , Redes Neurales de la Computación , Humanos , Animales , Neuronas/fisiología , Simulación por Computador , Red Nerviosa/fisiología , Dinámicas no Lineales
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda