Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 17549, 2024 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080344

RESUMEN

Virus‒host protein‒lncRNA interaction (VHPLI) predictions are critical for decoding the molecular mechanisms of viral pathogens and host immune processes. Although VHPLI interactions have been predicted in both plants and animals, they have not been extensively studied in viruses. For the first time, we propose a new deep learning-based approach that consists mainly of a convolutional neural network and bidirectional long and short-term memory network modules in combination with transfer learning named CBIL‒VHPLI to predict viral-host protein‒lncRNA interactions. The models were first trained on large and diverse datasets (including plants, animals, etc.). Protein sequence features were extracted using a k-mer method combined with the one-hot encoding and composition-transition-distribution (CTD) methods, and lncRNA sequence features were extracted using a k-mer method combined with the one-hot encoding and Z curve methods. The results obtained on three independent external validation datasets showed that the pre-trained CBIL‒VHPLI model performed the best with an accuracy of approximately 0.9. Pretraining was followed by conducting transfer learning on a viral protein-human lncRNA dataset, and the fine-tuning results showed that the accuracy of CBIL‒VHPLI was 0.946, which was significantly greater than that of the previous models. The final case study results showed that CBIL‒VHPLI achieved a prediction reproducibility rate of 91.6% for the RIP-Seq experimental screening results. This model was then used to predict the interactions between human lncRNA PIK3CD-AS2 and the nonstructural protein 1 (NS1) of the H5N1 virus, and RNA pull-down experiments were used to prove the prediction readiness of the model in terms of prediction. The source code of CBIL‒VHPLI and the datasets used in this work are available at https://github.com/Liu-Lab-Lnu/CBIL-VHPLI for academic usage.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Aprendizaje Automático , Proteínas Virales/metabolismo , Proteínas Virales/genética , Interacciones Huésped-Patógeno/genética , Aprendizaje Profundo , Redes Neurales de la Computación , Biología Computacional/métodos
2.
Mol Ther Nucleic Acids ; 35(2): 102187, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38706631

RESUMEN

Long non-coding RNAs (lncRNAs) are important factors involved in biological regulatory networks. Accurately predicting lncRNA-protein interactions (LPIs) is vital for clarifying lncRNA's functions and pathogenic mechanisms. Existing deep learning models have yet to yield satisfactory results in LPI prediction. Recently, graph autoencoders (GAEs) have seen rapid development, excelling in tasks like link prediction and node classification. We employed GAE technology for LPI prediction, devising the FMSRT-LPI model based on path masking and degree regression strategies and thereby achieving satisfactory outcomes. This represents the first known integration of path masking and degree regression strategies into the GAE framework for potential LPI inference. The effectiveness of our FMSRT-LPI model primarily relies on four key aspects. First, within the GAE framework, our model integrates multi-source relationships of lncRNAs and proteins with LPN's topological data. Second, the implemented masking strategy efficiently identifies LPN's key paths, reconstructs the network, and reduces the impact of redundant or incorrect data. Third, the integrated degree decoder balances degree and structural information, enhancing node representation. Fourth, the PolyLoss function we introduced is more appropriate for LPI prediction tasks. The results on multiple public datasets further demonstrate our model's potential in LPI prediction.

3.
Microrna ; 13(2): 155-165, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38591194

RESUMEN

BACKGROUND: Long non-coding RNA (lncRNA) plays a crucial role in various biological processes, and mutations or imbalances of lncRNAs can lead to several diseases, including cancer, Prader-Willi syndrome, autism, Alzheimer's disease, cartilage-hair hypoplasia, and hearing loss. Understanding lncRNA-protein interactions (LPIs) is vital for elucidating basic cellular processes, human diseases, viral replication, transcription, and plant pathogen resistance. Despite the development of several LPI calculation methods, predicting LPI remains challenging, with the selection of variables and deep learning structure being the focus of LPI research. METHODS: We propose a deep learning framework called AR-LPI, which extracts sequence and secondary structure features of proteins and lncRNAs. The framework utilizes an auto-encoder for feature extraction and employs SE-ResNet for prediction. Additionally, we apply transfer learning to the deep neural network SE-ResNet for predicting small-sample datasets. RESULTS: Through comprehensive experimental comparison, we demonstrate that the AR-LPI architecture performs better in LPI prediction. Specifically, the accuracy of AR-LPI increases by 2.86% to 94.52%, while the F-value of AR-LPI increases by 2.71% to 94.73%. CONCLUSION: Our experimental results show that the overall performance of AR-LPI is better than that of other LPI prediction tools.


Asunto(s)
Aprendizaje Profundo , ARN Largo no Codificante , ARN Largo no Codificante/genética , Humanos , Redes Neurales de la Computación , Biología Computacional/métodos
4.
BMC Bioinformatics ; 25(1): 108, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38475723

RESUMEN

RNA-protein interaction (RPI) is crucial to the life processes of diverse organisms. Various researchers have identified RPI through long-term and high-cost biological experiments. Although numerous machine learning and deep learning-based methods for predicting RPI currently exist, their robustness and generalizability have significant room for improvement. This study proposes LPI-MFF, an RPI prediction model based on multi-source information fusion, to address these issues. The LPI-MFF employed protein-protein interactions features, sequence features, secondary structure features, and physical and chemical properties as the information sources with the corresponding coding scheme, followed by the random forest algorithm for feature screening. Finally, all information was combined and a classification method based on convolutional neural networks is used. The experimental results of fivefold cross-validation demonstrated that the accuracy of LPI-MFF on RPI1807 and NPInter was 97.60% and 97.67%, respectively. In addition, the accuracy rate on the independent test set RPI1168 was 84.9%, and the accuracy rate on the Mus musculus dataset was 90.91%. Accordingly, LPI-MFF demonstrated greater robustness and generalization than other prevalent RPI prediction methods.


Asunto(s)
Aprendizaje Profundo , ARN Largo no Codificante , Animales , Ratones , ARN Largo no Codificante/química , Bosques Aleatorios , Redes Neurales de la Computación , Aprendizaje Automático , Biología Computacional/métodos
5.
Interdiscip Sci ; 16(2): 378-391, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38206558

RESUMEN

 Long noncoding RNAs (lncRNAs) have significant regulatory roles in gene expression. Interactions with proteins are one of the ways lncRNAs play their roles. Since experiments to determine lncRNA-protein interactions (LPIs) are expensive and time-consuming, many computational methods for predicting LPIs have been proposed as alternatives. In the LPIs prediction problem, there commonly exists the imbalance in the distribution of positive and negative samples. However, there are few existing methods that give specific consideration to this problem. In this paper, we proposed a new clustering-based LPIs prediction method using segmented k-mer frequencies and multi-space clustering (LPI-SKMSC). It was dedicated to handling the imbalance of positive and negative samples. We constructed segmented k-mer frequencies to obtain global and local features of lncRNA and protein sequences. Then, the multi-space clustering was applied to LPI-SKMSC. The convolutional neural network (CNN)-based encoders were used to map different features of a sample to different spaces. It used multiple spaces to jointly constrain the classification of samples. Finally, the distances between the output features of the encoder and the cluster center in each space were calculated. The sum of distances in all spaces was compared with the cluster radius to predict the LPIs. We performed cross-validation on 3 public datasets and LPI-SKMSC showed the best performance compared to other existing methods. Experimental results showed that LPI-SKMSC could predict LPIs more effectively when faced with imbalanced positive and negative samples. In addition, we illustrated that our model was better at uncovering potential lncRNA-protein interaction pairs.


Asunto(s)
Redes Neurales de la Computación , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Análisis por Conglomerados , Biología Computacional/métodos , Algoritmos , Proteínas/metabolismo , Proteínas/genética , Humanos
6.
Methods ; 220: 98-105, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37972912

RESUMEN

Many studies have shown that long-chain noncoding RNAs (lncRNAs) are involved in a variety of biological processes such as post-transcriptional gene regulation, splicing, and translation by combining with corresponding proteins. Predicting lncRNA-protein interactions is an effective approach to infer the functions of lncRNAs. The paper proposes a new computational model named LPI-IBWA. At first, LPI-IBWA uses similarity kernel fusion (SKF) to integrate various types of biological information to construct lncRNA and protein similarity networks. Then, a bounded matrix completion model and a weighted k-nearest known neighbors algorithm are utilized to update the initial sparse lncRNA-protein interaction matrix. Based on the updated lncRNA-protein interaction matrix, the lncRNA similarity network and the protein similarity network are integrated into a heterogeneous network. Finally, an improved Bi-Random walk algorithm is used to predict novel latent lncRNA-protein interactions. 5-fold cross-validation experiments on a benchmark dataset showed that the AUC and AUPR of LPI-IBWA reach 0.920 and 0.736, respectively, which are higher than those of other state-of-the-art methods. Furthermore, the experimental results of case studies on a novel dataset also illustrated that LPI-IBWA could efficiently predict potential lncRNA-protein interactions.


Asunto(s)
ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Algoritmos , Proteínas/metabolismo , Empalme del ARN , Biología Computacional/métodos
7.
Biochimie ; 214(Pt A): 123-140, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37353139

RESUMEN

Long non-coding RNAs (lncRNAs) are recently-discovered transcripts involved in gene expression regulation and associated with diseases. Despite the unprecedented molecular complexity of these transcripts, recent studies of the secondary and tertiary structure of lncRNAs are starting to reveal the principles of lncRNA structural organization, with important functional implications. It therefore starts to be possible to analyze lncRNA structures systematically. Here, using a set of prototypical and medically-relevant lncRNAs of known secondary structure, we specifically catalogue the distribution and structural environment of one of the first-identified and most frequently occurring non-canonical Watson-Crick interactions, the G·U base pair. We compare the properties of G·U base pairs in our set of lncRNAs to those of the G·U base pairs in other well-characterized transcripts, like rRNAs, tRNAs, ribozymes, and riboswitches. Furthermore, we discuss how G·U base pairs in these targets participate in establishing interactions with proteins or miRNAs, and how they enable lncRNA tertiary folding by forming intramolecular or metal-ion interactions. Finally, by identifying highly-G·U-enriched regions of yet unknown function in our target lncRNAs, we provide a new rationale for future experimental investigation of these motifs, which will help obtain a more comprehensive understanding of lncRNA functions and molecular mechanisms in the future.


Asunto(s)
ARN Largo no Codificante , Emparejamiento Base , ARN Largo no Codificante/genética , Conformación de Ácido Nucleico , ARN Ribosómico/química , ARN de Transferencia
8.
Int J Mol Sci ; 24(6)2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36982719

RESUMEN

Ethanol (EtOH) alters many cellular processes in yeast. An integrated view of different EtOH-tolerant phenotypes and their long noncoding RNAs (lncRNAs) is not yet available. Here, large-scale data integration showed the core EtOH-responsive pathways, lncRNAs, and triggers of higher (HT) and lower (LT) EtOH-tolerant phenotypes. LncRNAs act in a strain-specific manner in the EtOH stress response. Network and omics analyses revealed that cells prepare for stress relief by favoring activation of life-essential systems. Therefore, longevity, peroxisomal, energy, lipid, and RNA/protein metabolisms are the core processes that drive EtOH tolerance. By integrating omics, network analysis, and several other experiments, we showed how the HT and LT phenotypes may arise: (1) the divergence occurs after cell signaling reaches the longevity and peroxisomal pathways, with CTA1 and ROS playing key roles; (2) signals reaching essential ribosomal and RNA pathways via SUI2 enhance the divergence; (3) specific lipid metabolism pathways also act on phenotype-specific profiles; (4) HTs take greater advantage of degradation and membraneless structures to cope with EtOH stress; and (5) our EtOH stress-buffering model suggests that diauxic shift drives EtOH buffering through an energy burst, mainly in HTs. Finally, critical genes, pathways, and the first models including lncRNAs to describe nuances of EtOH tolerance are reported here.


Asunto(s)
ARN Largo no Codificante , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , ARN Largo no Codificante/genética , Etanol/farmacología , Etanol/metabolismo
9.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36515153

RESUMEN

Long noncoding RNA (lncRNA) is a kind of noncoding RNA with a length of more than 200 nucleotide units. Numerous research studies have proven that although lncRNAs cannot be directly translated into proteins, lncRNAs still play an important role in human growth processes by interacting with proteins. Since traditional biological experiments often require a lot of time and material costs to explore potential lncRNA-protein interactions (LPI), several computational models have been proposed for this task. In this study, we introduce a novel deep learning method known as combined graph auto-encoders (LPICGAE) to predict potential human LPIs. First, we apply a variational graph auto-encoder to learn the low dimensional representations from the high-dimensional features of lncRNAs and proteins. Then the graph auto-encoder is used to reconstruct the adjacency matrix for inferring potential interactions between lncRNAs and proteins. Finally, we minimize the loss of the two processes alternately to gain the final predicted interaction matrix. The result in 5-fold cross-validation experiments illustrates that our method achieves an average area under receiver operating characteristic curve of 0.974 and an average accuracy of 0.985, which is better than those of existing six state-of-the-art computational methods. We believe that LPICGAE can help researchers to gain more potential relationships between lncRNAs and proteins effectively.


Asunto(s)
Proteínas , ARN Largo no Codificante , Humanos , Biología Computacional/métodos , Proteínas/genética , Proteínas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Aprendizaje Profundo
10.
Comput Biol Chem ; 99: 107718, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35785626

RESUMEN

Long non-coding RNAs (LncRNAs) play important roles in a series of life activities, and they function primarily with proteins. The wet experimental-based methods in lncRNA-protein interactions (lncRPIs) study are time-consuming and expensive. In this study, we propose for the first time a novel feature fusion method, the LPI-CSFFR, to train and predict LncRPIs based on a Convolutional Neural Network (CNN) with feature reuse and serial fusion in sequences, secondary structures, and physicochemical properties of proteins and lncRNAs. The experimental results indicate that LPI-CSFFR achieves excellent performance on the datasets RPI1460 and RPI1807 with an accuracy of 83.7 % and 98.1 %, respectively. We further compare LPI-CSFFR with the state-of-the-art existing methods on the same benchmark datasets to evaluate the performance. In addition, to test the generalization performance of the model, we independently test sample pairs of five model organisms, where Mus musculus are the highest prediction accuracy of 99.5 %, and we find multiple hotspot proteins after constructing an interaction network. Finally, we test the predictive power of the LPI-CSFFR for sample pairs with unknown interactions. The results indicate that LPI-CSFFR is promising for predicting potential LncRPIs. The relevant source code and the data used in this study are available at https://github.com/JianjunTan-Beijing/LPI-CSFFR.


Asunto(s)
ARN Largo no Codificante , Animales , Biología Computacional/métodos , Ratones , Redes Neurales de la Computación , Proteínas/metabolismo , ARN Largo no Codificante/metabolismo , Programas Informáticos
11.
Genes (Basel) ; 12(11)2021 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-34828296

RESUMEN

Long noncoding RNA (lncRNA) plays a crucial role in many critical biological processes and participates in complex human diseases through interaction with proteins. Considering that identifying lncRNA-protein interactions through experimental methods is expensive and time-consuming, we propose a novel method based on deep learning that combines raw sequence composition features, hand-designed features and structure features, called LGFC-CNN, to predict lncRNA-protein interactions. The two sequence preprocessing methods and CNN modules (GloCNN and LocCNN) are utilized to extract the raw sequence global and local features. Meanwhile, we select hand-designed features by comparing the predictive effect of different lncRNA and protein features combinations. Furthermore, we obtain the structure features and unifying the dimensions through Fourier transform. In the end, the four types of features are integrated to comprehensively predict the lncRNA-protein interactions. Compared with other state-of-the-art methods on three lncRNA-protein interaction datasets, LGFC-CNN achieves the best performance with an accuracy of 94.14%, on RPI21850; an accuracy of 92.94%, on RPI7317; and an accuracy of 98.19% on RPI1847. The results show that our LGFC-CNN can effectively predict the lncRNA-protein interactions by combining raw sequence composition features, hand-designed features and structure features.


Asunto(s)
Aprendizaje Profundo , Redes Reguladoras de Genes/fisiología , Mapas de Interacción de Proteínas/fisiología , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Biología Computacional/instrumentación , Biología Computacional/métodos , Conjuntos de Datos como Asunto , Humanos , Redes Neurales de la Computación , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética
12.
Front Bioeng Biotechnol ; 9: 647113, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33718346

RESUMEN

The long non-coding RNA (lncRNA)-protein interaction plays an important role in the post-transcriptional gene regulation, such as RNA splicing, translation, signaling, and the development of complex diseases. The related research on the prediction of lncRNA-protein interaction relationship is beneficial in the excavation and the discovery of the mechanism of lncRNA function and action occurrence, which are important. Traditional experimental methods for detecting lncRNA-protein interactions are expensive and time-consuming. Therefore, computational methods provide many effective strategies to deal with this problem. In recent years, most computational methods only use the information of the lncRNA-lncRNA or the protein-protein similarity and cannot fully capture all features to identify their interactions. In this paper, we propose a novel computational model for the lncRNA-protein prediction on the basis of machine learning methods. First, a feature method is proposed for representing the information of the network topological properties of lncRNA and protein interactions. The basic composition feature information and evolutionary information based on protein, the lncRNA sequence feature information, and the lncRNA expression profile information are extracted. Finally, the above feature information is fused, and the optimized feature vector is used with the recursive feature elimination algorithm. The optimized feature vectors are input to the support vector machine (SVM) model. Experimental results show that the proposed method has good effectiveness and accuracy in the lncRNA-protein interaction prediction.

13.
Int J Mol Sci ; 22(2)2021 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-33435206

RESUMEN

Over the past decades, research on cancer biology has focused on the involvement of protein-coding genes in cancer development. Long noncoding RNAs (lncRNAs), which are transcripts longer than 200 nucleotides that lack protein-coding potential, are an important class of RNA molecules that are involved in a variety of biological functions. Although the functions of a majority of lncRNAs have yet to be clarified, some lncRNAs have been shown to be associated with human diseases such as cancer. LncRNAs have been shown to contribute to many important cancer phenotypes through their interactions with other cellular macromolecules including DNA, protein and RNA. Here we describe the literature regarding the biogenesis and features of lncRNAs. We also present an overview of the current knowledge regarding the roles of lncRNAs in cancer from the view of various aspects of cellular homeostasis, including proliferation, survival, migration and genomic stability. Furthermore, we discuss the methodologies used to identify the function of lncRNAs in cancer development and tumorigenesis. Better understanding of the molecular mechanisms involving lncRNA functions in cancer is critical for the development of diagnostic and therapeutic strategies against tumorigenesis.


Asunto(s)
Neoplasias/genética , ARN Largo no Codificante/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/metabolismo
14.
Anal Biochem ; 601: 113767, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32454029

RESUMEN

Long noncoding RNAs (lncRNAs) play critical roles in many pathological and biological processes, such as post-transcription, cell differentiation and gene regulation. Increasingly more studies have shown that lncRNAs function through mainly interactions with specific RNA binding proteins (RBPs). However, experimental identification of potential lncRNA-protein interactions is costly and time-consuming. In this work, we propose a novel convolutional neural network-based method with the copy-padding trick (named LPI-CNNCP) to predict lncRNA-protein interactions. The copy-padding trick of the LPI-CNNCP convert the protein/RNA sequences with variable-length into the fixed-length sequences, thus enabling the construction of the CNN model. A high-order one-hot encoding is also applied to transform the protein/RNA sequences into image-like inputs for capturing the dependencies among amino acids (or nucleotides). In the end, these encoded protein/RNA sequences are feed into a CNN to predict the lncRNA-protein interactions. Compared with other state-of-the-art methods in 10-fold cross-validation (10CV) test, LPI-CNNCP shows the best performance. Results in the independent test demonstrate that our LPI-CNNCP can effectively predict the potential lncRNA-protein interactions. We also compared the copy-padding trick with two other existing tricks (i.e., zero-padding and cropping), and the results show that our copy-padding rick outperforms the zero-padding and cropping tricks on predicting lncRNA-protein interactions. The source code of LPI-CNNCP and the datasets used in this work are available at https://github.com/NWPU-903PR/LPI-CNNCP for academic users.


Asunto(s)
Redes Neurales de la Computación , ARN Largo no Codificante/química , Proteínas de Unión al ARN/química , Secuencia de Aminoácidos , Humanos
15.
Front Genet ; 10: 772, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31507635

RESUMEN

Polycystic ovary syndrome (PCOS) is a common metabolic and reproductive disorder with an increasing risk for type 2 diabetes. Insulin resistance is a common feature of women with PCOS, but the underlying molecular mechanism remains unclear. This study aimed to screen critical long non-coding RNAs (lncRNAs) that might play pivotal roles in insulin resistance, which could provide candidate biomarkers and potential therapeutic targets for PCOS. Gene expression profiles of the skeletal muscle in patients with PCOS accompanied by insulin resistance and healthy patients were obtained from the publicly available Gene Expression Omnibus (GEO) database. A global triple network including RNA-binding protein, mRNA, and lncRNAs was constructed based on the data from starBase. Then, we extracted an insulin resistance-associated lncRNA-mRNA network (IRLMN) by integrating the data from starBase and GEO. We also performed a weighted gene co-expression network analysis (WGCNA) on the differentially expressed genes between the women with and without PCOS, to identify hub lncRNAs. Additionally, the findings of key lncRNAs were examined in an independent GEO dataset. The expression level of lncRNA RP11-151A6.4 in ovarian granulosa cells was increased in patients with PCOS compared with that in control women. Levels were also increased in PCOS patients with higher BMI, hyperinsulinemia, and higher HOMA-IR values. As a result, RP11-151A6.4 was identified as a hub lncRNA based on IRLMN and WGCNA and was highly expressed in ovarian granulosa cells, skeletal muscle, and subcutaneous and omental adipose tissues of patients with insulin resistance. This study showed the differences between lncRNA and mRNA profiles from healthy women and women with PCOS and insulin resistance. Here, we demonstrated that RP11-151A6.4 might play a vital role in insulin resistance, androgen excess, and adipose dysfunction in patients with PCOS. Further study concerning RP11-151A6.4 could elucidate the underlying mechanisms of insulin resistance.

16.
Front Genet ; 9: 716, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30697228

RESUMEN

Long non-coding RNAs (lncRNAs) constitute a large class of transcribed RNA molecules. They have a characteristic length of more than 200 nucleotides which do not encode proteins. They play an important role in regulating gene expression by interacting with the homologous RNA-binding proteins. Due to the laborious and time-consuming nature of wet experimental methods, more researchers should pay great attention to computational approaches for the prediction of lncRNA-protein interaction (LPI). An in-depth literature review in the state-of-the-art in silico investigations, leads to the conclusion that there is still room for improving the accuracy and velocity. This paper propose a novel method for identifying LPI by employing Kernel Ridge Regression, based on Fast Kernel Learning (LPI-FKLKRR). This approach, uses four distinct similarity measures for lncRNA and protein space, respectively. It is remarkable, that we extract Gene Ontology (GO) with proteins, in order to improve the quality of information in protein space. The process of heterogeneous kernels integration, applies Fast Kernel Learning (FastKL) to deal with weight optimization. The extrapolation model is obtained by gaining the ultimate prediction associations, after using Kernel Ridge Regression (KRR). Experimental outcomes show that the ability of modeling with LPI-FKLKRR has extraordinary performance compared with LPI prediction schemes. On benchmark dataset, it has been observed that the best Area Under Precision Recall Curve (AUPR) of 0.6950 is obtained by our proposed model LPI-FKLKRR, which outperforms the integrated LPLNP (AUPR: 0.4584), RWR (AUPR: 0.2827), CF (AUPR: 0.2357), LPIHN (AUPR: 0.2299), and LPBNI (AUPR: 0.3302). Also, combined with the experimental results of a case study on a novel dataset, it is anticipated that LPI-FKLKRR will be a useful tool for LPI prediction.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA