Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genomics Proteomics Bioinformatics ; 21(5): 913-925, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37001856

RESUMO

Protein structure prediction is an interdisciplinary research topic that has attracted researchers from multiple fields, including biochemistry, medicine, physics, mathematics, and computer science. These researchers adopt various research paradigms to attack the same structure prediction problem: biochemists and physicists attempt to reveal the principles governing protein folding; mathematicians, especially statisticians, usually start from assuming a probability distribution of protein structures given a target sequence and then find the most likely structure, while computer scientists formulate protein structure prediction as an optimization problem - finding the structural conformation with the lowest energy or minimizing the difference between predicted structure and native structure. These research paradigms fall into the two statistical modeling cultures proposed by Leo Breiman, namely, data modeling and algorithmic modeling. Recently, we have also witnessed the great success of deep learning in protein structure prediction. In this review, we present a survey of the efforts for protein structure prediction. We compare the research paradigms adopted by researchers from different fields, with an emphasis on the shift of research paradigms in the era of deep learning. In short, the algorithmic modeling techniques, especially deep neural networks, have considerably improved the accuracy of protein structure prediction; however, theories interpreting the neural networks and knowledge on protein folding are still highly desired.


Assuntos
Algoritmos , Proteínas , Conformação Proteica , Proteínas/química , Redes Neurais de Computação , Dobramento de Proteína , Biologia Computacional/métodos
2.
J Comput Biol ; 29(2): 92-105, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35073170

RESUMO

Template-based modeling (TBM), including homology modeling and protein threading, is one of the most reliable techniques for protein structure prediction. It predicts protein structure by building an alignment between the query sequence under prediction and the templates with solved structures. However, it is still very challenging to build the optimal sequence-template alignment, especially when only distantly related templates are available. Here we report a novel deep learning approach ProALIGN that can predict much more accurate sequence-template alignment. Like protein sequences consisting of sequence motifs, protein alignments are also composed of frequently occurring alignment motifs with characteristic patterns. Alignment motifs are context-specific as their characteristic patterns are tightly related to sequence contexts of the aligned regions. Inspired by this observation, we represent a protein alignment as a binary matrix (in which 1 denotes an aligned residue pair) and then use a deep convolutional neural network to predict the optimal alignment from the query protein and its template. The trained neural network implicitly but effectively encodes an alignment scoring function, which reduces inaccuracies in the handcrafted scoring functions widely used by the current threading approaches. For a query protein and a template, we apply the neural network to directly infer likelihoods of all possible residue pairs in their entirety, which could effectively consider the correlations among multiple residues. We further construct the alignment with maximum likelihood, and finally build a structure model according to the alignment. Tested on three independent data sets with a total of 6688 protein alignment targets and 80 CASP13 TBM targets, our method achieved much better alignments and 3D structure models than the existing methods, including HHpred, CNFpred, CEthreader, and DeepThreader. These results clearly demonstrate the effectiveness of exploiting the context-specific alignment motifs by deep learning for protein threading.


Assuntos
Aprendizado Profundo , Proteínas/química , Alinhamento de Sequência/estatística & dados numéricos , Algoritmos , Motivos de Aminoácidos , Sequência de Aminoácidos , Biologia Computacional , Modelos Moleculares , Redes Neurais de Computação , Conformação Proteica , Proteínas/genética , Análise de Sequência de Proteína/estatística & dados numéricos , Software
3.
BMC Bioinformatics ; 22(1): 439, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34525939

RESUMO

BACKGROUND: Accurate prediction of protein tertiary structures is highly desired as the knowledge of protein structures provides invaluable insights into protein functions. We have designed two approaches to protein structure prediction, including a template-based modeling approach (called ProALIGN) and an ab initio prediction approach (called ProFOLD). Briefly speaking, ProALIGN aligns a target protein with templates through exploiting the patterns of context-specific alignment motifs and then builds the final structure with reference to the homologous templates. In contrast, ProFOLD uses an end-to-end neural network to estimate inter-residue distances of target proteins and builds structures that satisfy these distance constraints. These two approaches emphasize different characteristics of target proteins: ProALIGN exploits structure information of homologous templates of target proteins while ProFOLD exploits the co-evolutionary information carried by homologous protein sequences. Recent progress has shown that the combination of template-based modeling and ab initio approaches is promising. RESULTS: In the study, we present FALCON2, a web server that integrates ProALIGN and ProFOLD to provide high-quality protein structure prediction service. For a target protein, FALCON2 executes ProALIGN and ProFOLD simultaneously to predict possible structures and selects the most likely one as the final prediction result. We evaluated FALCON2 on widely-used benchmarks, including 104 CASP13 (the 13th Critical Assessment of protein Structure Prediction) targets and 91 CASP14 targets. In-depth examination suggests that when high-quality templates are available, ProALIGN is superior to ProFOLD and in other cases, ProFOLD shows better performance. By integrating these two approaches with different emphasis, FALCON2 server outperforms the two individual approaches and also achieves state-of-the-art performance compared with existing approaches. CONCLUSIONS: By integrating template-based modeling and ab initio approaches, FALCON2 provides an easy-to-use and high-quality protein structure prediction service for the community and we expect it to enable insights into a deep understanding of protein functions.


Assuntos
Redes Neurais de Computação , Proteínas , Sequência de Aminoácidos , Computadores , Conformação Proteica , Software
4.
Nat Commun ; 12(1): 2535, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33953201

RESUMO

Residue co-evolution has become the primary principle for estimating inter-residue distances of a protein, which are crucially important for predicting protein structure. Most existing approaches adopt an indirect strategy, i.e., inferring residue co-evolution based on some hand-crafted features, say, a covariance matrix, calculated from multiple sequence alignment (MSA) of target protein. This indirect strategy, however, cannot fully exploit the information carried by MSA. Here, we report an end-to-end deep neural network, CopulaNet, to estimate residue co-evolution directly from MSA. The key elements of CopulaNet include: (i) an encoder to model context-specific mutation for each residue; (ii) an aggregator to model residue co-evolution, and thereafter estimate inter-residue distances. Using CASP13 (the 13th Critical Assessment of Protein Structure Prediction) target proteins as representatives, we demonstrate that CopulaNet can predict protein structure with improved accuracy and efficiency. This study represents a step toward improved end-to-end prediction of inter-residue distances and protein tertiary structures.


Assuntos
Aprendizado de Máquina , Proteínas/química , Alinhamento de Sequência , Caspases/química , Biologia Computacional , Humanos , Modelos Moleculares , Mutação , Redes Neurais de Computação , Estrutura Terciária de Proteína , Proteínas/genética
5.
BMC Bioinformatics ; 21(1): 503, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-33153432

RESUMO

BACKGROUND: The formation of contacts among protein secondary structure elements (SSEs) is an important step in protein folding as it determines topology of protein tertiary structure; hence, inferring inter-SSE contacts is crucial to protein structure prediction. One of the existing strategies infers inter-SSE contacts directly from the predicted possibilities of inter-residue contacts without any preprocessing, and thus suffers from the excessive noises existing in the predicted inter-residue contacts. Another strategy defines SSEs based on protein secondary structure prediction first, and then judges whether each candidate SSE pair could form contact or not. However, it is difficult to accurately determine boundary of SSEs due to the errors in secondary structure prediction. The incorrectly-deduced SSEs definitely hinder subsequent prediction of the contacts among them. RESULTS: We here report an accurate approach to infer the inter-SSE contacts (thus called as ISSEC) using the deep object detection technique. The design of ISSEC is based on the observation that, in the inter-residue contact map, the contacting SSEs usually form rectangle regions with characteristic patterns. Therefore, ISSEC infers inter-SSE contacts through detecting such rectangle regions. Unlike the existing approach directly using the predicted probabilities of inter-residue contact, ISSEC applies the deep convolution technique to extract high-level features from the inter-residue contacts. More importantly, ISSEC does not rely on the pre-defined SSEs. Instead, ISSEC enumerates multiple candidate rectangle regions in the predicted inter-residue contact map, and for each region, ISSEC calculates a confidence score to measure whether it has characteristic patterns or not. ISSEC employs greedy strategy to select non-overlapping regions with high confidence score, and finally infers inter-SSE contacts according to these regions. CONCLUSIONS: Comprehensive experimental results suggested that ISSEC outperformed the state-of-the-art approaches in predicting inter-SSE contacts. We further demonstrated the successful applications of ISSEC to improve prediction of both inter-residue contacts and tertiary structure as well.


Assuntos
Algoritmos , Proteínas/química , Bases de Dados de Proteínas , Proteínas de Membrana/química , Conformação Proteica em Folha beta , Estrutura Secundária de Proteína
6.
BMC Bioinformatics ; 20(Suppl 3): 135, 2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-30925867

RESUMO

BACKGROUND: The ab initio approaches to protein structure prediction usually employ the Monte Carlo technique to search the structural conformation that has the lowest energy. However, the widely-used energy functions are usually ineffective for conformation search. How to construct an effective energy function remains a challenging task. RESULTS: Here, we present a framework to construct effective energy functions for protein structure prediction. Unlike existing energy functions only requiring the native structure to be the lowest one, we attempt to maximize the attraction-basin where the native structure lies in the energy landscape. The underlying rationale is that each energy function determines a specific energy landscape together with a native attraction-basin, and the larger the attraction-basin is, the more likely for the Monte Carlo search procedure to find the native structure. Following this rationale, we constructed effective energy functions as follows: i) To explore the native attraction-basin determined by a certain energy function, we performed reverse Monte Carlo sampling starting from the native structure, identifying the structural conformations on the edge of attraction-basin. ii) To broaden the native attraction-basin, we smoothened the edge points of attraction-basin through tuning weights of energy terms, thus acquiring an improved energy function. Our framework alternates the broadening attraction-basin and reverse sampling steps (thus called BARS) until the native attraction-basin is sufficiently large. We present extensive experimental results to show that using the BARS framework, the constructed energy functions could greatly facilitate protein structure prediction in improving the quality of predicted structures and speeding up conformation search. CONCLUSION: Using the BARS framework, we constructed effective energy functions for protein structure prediction, which could improve the quality of predicted structures and speed up conformation search as well.


Assuntos
Biologia Computacional/métodos , Método de Monte Carlo , Proteínas/química , Algoritmos , Bases de Dados de Proteínas , Conformação Proteica , Termodinâmica
7.
Bioinformatics ; 33(23): 3749-3757, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28961795

RESUMO

MOTIVATION: Accurate recognition of protein fold types is a key step for template-based prediction of protein structures. The existing approaches to fold recognition mainly exploit the features derived from alignments of query protein against templates. These approaches have been shown to be successful for fold recognition at family level, but usually failed at superfamily/fold levels. To overcome this limitation, one of the key points is to explore more structurally informative features of proteins. Although residue-residue contacts carry abundant structural information, how to thoroughly exploit these information for fold recognition still remains a challenge. RESULTS: In this study, we present an approach (called DeepFR) to improve fold recognition at superfamily/fold levels. The basic idea of our approach is to extract fold-specific features from predicted residue-residue contacts of proteins using deep convolutional neural network (DCNN) technique. Based on these fold-specific features, we calculated similarity between query protein and templates, and then assigned query protein with fold type of the most similar template. DCNN has showed excellent performance in image feature extraction and image recognition; the rational underlying the application of DCNN for fold recognition is that contact likelihood maps are essentially analogy to images, as they both display compositional hierarchy. Experimental results on the LINDAHL dataset suggest that even using the extracted fold-specific features alone, our approach achieved success rate comparable to the state-of-the-art approaches. When further combining these features with traditional alignment-related features, the success rate of our approach increased to 92.3%, 82.5% and 78.8% at family, superfamily and fold levels, respectively, which is about 18% higher than the state-of-the-art approach at fold level, 6% higher at superfamily level and 1% higher at family level. An independent assessment on SCOP_TEST dataset showed consistent performance improvement, indicating robustness of our approach. Furthermore, bi-clustering results of the extracted features are compatible with fold hierarchy of proteins, implying that these features are fold-specific. Together, these results suggest that the features extracted from predicted contacts are orthogonal to alignment-related features, and the combination of them could greatly facilitate fold recognition at superfamily/fold levels and template-based prediction of protein structures. AVAILABILITY AND IMPLEMENTATION: Source code of DeepFR is freely available through https://github.com/zhujianwei31415/deepfr, and a web server is available through http://protein.ict.ac.cn/deepfr. CONTACT: zheng@itp.ac.cn or dbu@ict.ac.cn. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Dobramento de Proteína , Algoritmos , Redes Neurais de Computação , Proteínas/química , Software
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...