Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Proteins ; 91(12): 1539-1549, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37920879

RESUMO

Computing protein structure from amino acid sequence information has been a long-standing grand challenge. Critical assessment of structure prediction (CASP) conducts community experiments aimed at advancing solutions to this and related problems. Experiments are conducted every 2 years. The 2020 experiment (CASP14) saw major progress, with the second generation of deep learning methods delivering accuracy comparable with experiment for many single proteins. There is an expectation that these methods will have much wider application in computational structural biology. Here we summarize results from the most recent experiment, CASP15, in 2022, with an emphasis on new deep learning-driven progress. Other papers in this special issue of proteins provide more detailed analysis. For single protein structures, the AlphaFold2 deep learning method is still superior to other approaches, but there are two points of note. First, although AlphaFold2 was the core of all the most successful methods, there was a wide variety of implementation and combination with other methods. Second, using the standard AlphaFold2 protocol and default parameters only produces the highest quality result for about two thirds of the targets, and more extensive sampling is required for the others. The major advance in this CASP is the enormous increase in the accuracy of computed protein complexes, achieved by the use of deep learning methods, although overall these do not fully match the performance for single proteins. Here too, AlphaFold2 based method perform best, and again more extensive sampling than the defaults is often required. Also of note are the encouraging early results on the use of deep learning to compute ensembles of macromolecular structures. Critically for the usability of computed structures, for both single proteins and protein complexes, deep learning derived estimates of both local and global accuracy are of high quality, however the estimates in interface regions are slightly less reliable. CASP15 also included computation of RNA structures for the first time. Here, the classical approaches produced better agreement with experiment than the new deep learning ones, and accuracy is limited. Also, for the first time, CASP included the computation of protein-ligand complexes, an area of special interest for drug design. Here too, classical methods were still superior to deep learning ones. Many new approaches were discussed at the CASP conference, and it is clear methods will continue to advance.


Assuntos
Biologia Computacional , Proteínas , Conformação Proteica , Modelos Moleculares , Proteínas/química , Sequência de Aminoácidos , Biologia Computacional/métodos
2.
Proteins ; 91(12): 1571-1599, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37493353

RESUMO

We present an in-depth analysis of selected CASP15 targets, focusing on their biological and functional significance. The authors of the structures identify and discuss key protein features and evaluate how effectively these aspects were captured in the submitted predictions. While the overall ability to predict three-dimensional protein structures continues to impress, reproducing uncommon features not previously observed in experimental structures is still a challenge. Furthermore, instances with conformational flexibility and large multimeric complexes highlight the need for novel scoring strategies to better emphasize biologically relevant structural regions. Looking ahead, closer integration of computational and experimental techniques will play a key role in determining the next challenges to be unraveled in the field of structural molecular biology.


Assuntos
Biologia Computacional , Proteínas , Conformação Proteica , Modelos Moleculares , Biologia Computacional/métodos , Proteínas/química
3.
Proteins ; 91(12): 1550-1557, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37306011

RESUMO

Prediction categories in the Critical Assessment of Structure Prediction (CASP) experiments change with the need to address specific problems in structure modeling. In CASP15, four new prediction categories were introduced: RNA structure, ligand-protein complexes, accuracy of oligomeric structures and their interfaces, and ensembles of alternative conformations. This paper lists technical specifications for these categories and describes their integration in the CASP data management system.


Assuntos
Biologia Computacional , Proteínas , Conformação Proteica , Proteínas/química , Modelos Moleculares , Ligantes
4.
Nat Commun ; 14(1): 2366, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37185902

RESUMO

The Papain-like protease (PLpro) is a domain of a multi-functional, non-structural protein 3 of coronaviruses. PLpro cleaves viral polyproteins and posttranslational conjugates with poly-ubiquitin and protective ISG15, composed of two ubiquitin-like (UBL) domains. Across coronaviruses, PLpro showed divergent selectivity for recognition and cleavage of posttranslational conjugates despite sequence conservation. We show that SARS-CoV-2 PLpro binds human ISG15 and K48-linked di-ubiquitin (K48-Ub2) with nanomolar affinity and detect alternate weaker-binding modes. Crystal structures of untethered PLpro complexes with ISG15 and K48-Ub2 combined with solution NMR and cross-linking mass spectrometry revealed how the two domains of ISG15 or K48-Ub2 are differently utilized in interactions with PLpro. Analysis of protein interface energetics predicted differential binding stabilities of the two UBL/Ub domains that were validated experimentally. We emphasize how substrate recognition can be tuned to cleave specifically ISG15 or K48-Ub2 modifications while retaining capacity to cleave mono-Ub conjugates. These results highlight alternative druggable surfaces that would inhibit PLpro function.


Assuntos
COVID-19 , SARS-CoV-2 , Ubiquitina , Humanos , Citocinas/metabolismo , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , SARS-CoV-2/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo
5.
bioRxiv ; 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-35547846

RESUMO

The Papain-like protease (PLpro) is a domain of a multi-functional, non-structural protein 3 of coronaviruses. PLpro cleaves viral polyproteins and posttranslational conjugates with poly-ubiquitin and protective ISG15, composed of two ubiquitin-like (UBL) domains. Across coronaviruses, PLpro showed divergent selectivity for recognition and cleavage of posttranslational conjugates despite sequence conservation. We show that SARS-CoV-2 PLpro binds human ISG15 and K48-linked di-ubiquitin (K48-Ub 2 ) with nanomolar affinity and detect alternate weaker-binding modes. Crystal structures of untethered PLpro complexes with ISG15 and K48-Ub 2 combined with solution NMR and cross-linking mass spectrometry revealed how the two domains of ISG15 or K48-Ub 2 are differently utilized in interactions with PLpro. Analysis of protein interface energetics predicted differential binding stabilities of the two UBL/Ub domains that were validated experimentally. We emphasize how substrate recognition can be tuned to cleave specifically ISG15 or K48-Ub 2 modifications while retaining capacity to cleave mono-Ub conjugates. These results highlight alternative druggable surfaces that would inhibit PLpro function.

6.
Pac Symp Biocomput ; 27: 1-9, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34890131

RESUMO

The last few years mark dramatic improvements in modeling of protein structure. Progress was initially due to breakthroughs in residue-residue contact prediction, first with global statistical models and later with deep learning. These advancements were then followed by an even broader application of the deep learning techniques to the protein structure modeling itself, first using Convolutional Neural Networks (CNNs) and then switching to Natural Language Processing (NLP), including Attention models, and to Geometric Deep Learning (GDL). The accuracy of protein structure models generated with current state-of-the-art methods rivals that of experimental structures, while models themselves are used to solve structures or to make them more accurate.Looking at the near future of machine learning applications in structural biology, we ask the following questions: Which specific problems should we expect to be solved next? Which new methods will prove to be the most effective? Which actions are likely to stimulate further progress the most? In addressing these questions, we invite the 2022 PSB attendees to actively participate in session discussions.The AI-driven Advances in Modeling of Protein Structure session includes five papers specifically dedicated to:Evaluating the significance of training data selection in machine learning.Geometric pattern transferability, from protein self-interactions to protein-ligand interactions.Supervised versus unsupervised sequence to contact learning, using attention models.Side chain packing using SE(3) transformers.Feature detection in electrostatic representations of ligand binding sites.


Assuntos
Aprendizado Profundo , Biologia Computacional , Humanos , Aprendizado de Máquina , Redes Neurais de Computação , Proteínas
7.
Proteins ; 89(12): 1607-1617, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34533838

RESUMO

Critical assessment of structure prediction (CASP) is a community experiment to advance methods of computing three-dimensional protein structure from amino acid sequence. Core components are rigorous blind testing of methods and evaluation of the results by independent assessors. In the most recent experiment (CASP14), deep-learning methods from one research group consistently delivered computed structures rivaling the corresponding experimental ones in accuracy. In this sense, the results represent a solution to the classical protein-folding problem, at least for single proteins. The models have already been shown to be capable of providing solutions for problematic crystal structures, and there are broad implications for the rest of structural biology. Other research groups also substantially improved performance. Here, we describe these results and outline some of the many implications. Other related areas of CASP, including modeling of protein complexes, structure refinement, estimation of model accuracy, and prediction of inter-residue contacts and distances, are also described.


Assuntos
Conformação Proteica , Dobramento de Proteína , Proteínas , Software , Sequência de Aminoácidos , Biologia Computacional , Modelos Estatísticos , Simulação de Dinâmica Molecular , Proteínas/química , Proteínas/metabolismo , Análise de Sequência de Proteína
8.
Proteins ; 89(12): 1959-1976, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34559429

RESUMO

NMR studies can provide unique information about protein conformations in solution. In CASP14, three reference structures provided by solution NMR methods were available (T1027, T1029, and T1055), as well as a fourth data set of NMR-derived contacts for an integral membrane protein (T1088). For the three targets with NMR-based structures, the best prediction results ranged from very good (GDT_TS = 0.90, for T1055) to poor (GDT_TS = 0.47, for T1029). We explored the basis of these results by comparing all CASP14 prediction models against experimental NMR data. For T1027, NMR data reveal extensive internal dynamics, presenting a unique challenge for protein structure prediction methods. The analysis of T1029 motivated exploration of a novel method of "inverse structure determination," in which an AlphaFold2 model was used to guide NMR data analysis. NMR data provided to CASP predictor groups for target T1088, a 238-residue integral membrane porin, was also used to assess several NMR-assisted prediction methods. Most groups involved in this exercise generated similar beta-barrel models, with good agreement with the experimental data. However, as was also observed in CASP13, some pure prediction groups that did not use any NMR data generated models for T1088 that better fit the NMR data than the models generated using these experimental data. These results demonstrate the remarkable power of modern methods to predict structures of proteins with accuracies rivaling solution NMR structures, and that it is now possible to reliably use prediction models to guide and complement experimental NMR data analysis.


Assuntos
Espectroscopia de Ressonância Magnética/métodos , Proteínas de Membrana , Modelos Moleculares , Conformação Proteica , Software , Biologia Computacional , Aprendizado de Máquina , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Dobramento de Proteína , Análise de Sequência de Proteína
9.
Proteins ; 89(12): 1647-1672, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34561912

RESUMO

The biological and functional significance of selected Critical Assessment of Techniques for Protein Structure Prediction 14 (CASP14) targets are described by the authors of the structures. The authors highlight the most relevant features of the target proteins and discuss how well these features were reproduced in the respective submitted predictions. The overall ability to predict three-dimensional structures of proteins has improved remarkably in CASP14, and many difficult targets were modeled with impressive accuracy. For the first time in the history of CASP, the experimentalists not only highlighted that computational models can accurately reproduce the most critical structural features observed in their targets, but also envisaged that models could serve as a guidance for further studies of biologically-relevant properties of proteins.


Assuntos
Modelos Moleculares , Conformação Proteica , Proteínas/química , Software , Sequência de Aminoácidos , Biologia Computacional , Microscopia Crioeletrônica , Cristalografia por Raios X , Análise de Sequência de Proteína
10.
Proteins ; 89(12): 1987-1996, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34462960

RESUMO

Critical Assessment of Structure Prediction (CASP) is an organization aimed at advancing the state of the art in computing protein structure from sequence. In the spring of 2020, CASP launched a community project to compute the structures of the most structurally challenging proteins coded for in the SARS-CoV-2 genome. Forty-seven research groups submitted over 3000 three-dimensional models and 700 sets of accuracy estimates on 10 proteins. The resulting models were released to the public. CASP community members also worked together to provide estimates of local and global accuracy and identify structure-based domain boundaries for some proteins. Subsequently, two of these structures (ORF3a and ORF8) have been solved experimentally, allowing assessment of both model quality and the accuracy estimates. Models from the AlphaFold2 group were found to have good agreement with the experimental structures, with main chain GDT_TS accuracy scores ranging from 63 (a correct topology) to 87 (competitive with experiment).


Assuntos
SARS-CoV-2/química , Proteínas Virais/química , COVID-19/virologia , Genoma Viral , Humanos , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , SARS-CoV-2/genética , Proteínas Virais/genética , Proteínas Viroporinas/química , Proteínas Viroporinas/genética
12.
Proteins ; 87(12): 1283-1297, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31569265

RESUMO

With the advance of experimental procedures obtaining chemical crosslinking information is becoming a fast and routine practice. Information on crosslinks can greatly enhance the accuracy of protein structure modeling. Here, we review the current state of the art in modeling protein structures with the assistance of experimentally determined chemical crosslinks within the framework of the 13th meeting of Critical Assessment of Structure Prediction approaches. This largest-to-date blind assessment reveals benefits of using data assistance in difficult to model protein structure prediction cases. However, in a broader context, it also suggests that with the unprecedented advance in accuracy to predict contacts in recent years, experimental crosslinks will be useful only if their specificity and accuracy further improved and they are better integrated into computational workflows.


Assuntos
Biologia Computacional/métodos , Reagentes de Ligações Cruzadas/química , Modelos Moleculares , Conformação Proteica , Proteínas/química , Algoritmos , Cromatografia Líquida , Modelos Químicos , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
13.
Proteins ; 87(12): 1058-1068, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31587357

RESUMO

The accuracy of sequence-based tertiary contact predictions was assessed in a blind prediction experiment at the CASP13 meeting. After 4 years of significant improvements in prediction accuracy, another dramatic advance has taken place since CASP12 was held 2 years ago. The precision of predicting the top L/5 contacts in the free modeling category, where L is the corresponding length of the protein in residues, has exceeded 70%. As a comparison, the best-performing group at CASP12 with a 47% precision would have finished below the top 1/3 of the CASP13 groups. Extensively trained deep neural network approaches dominate the top performing algorithms, which appear to efficiently integrate information on coevolving residues and interacting fragments or possibly utilize memories of sequence similarities and sometimes can deliver accurate results even in the absence of virtually any target specific evolutionary information. If the current performance is evaluated by F-score on L contacts, it stands around 24% right now, which, despite the tremendous impact and advance in improving its utility for structure modeling, also suggests that there is much room left for further improvement.


Assuntos
Biologia Computacional/métodos , Congressos como Assunto/estatística & dados numéricos , Conformação Proteica , Proteínas/química , Análise de Sequência de Proteína/métodos , Algoritmos , Congressos como Assunto/normas , Cristalografia por Raios X , Entropia , Humanos , Modelos Moleculares , Reprodutibilidade dos Testes
14.
Proteins ; 87(12): 1011-1020, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31589781

RESUMO

CASP (critical assessment of structure prediction) assesses the state of the art in modeling protein structure from amino acid sequence. The most recent experiment (CASP13 held in 2018) saw dramatic progress in structure modeling without use of structural templates (historically "ab initio" modeling). Progress was driven by the successful application of deep learning techniques to predict inter-residue distances. In turn, these results drove dramatic improvements in three-dimensional structure accuracy: With the proviso that there are an adequate number of sequences known for the protein family, the new methods essentially solve the long-standing problem of predicting the fold topology of monomeric proteins. Further, the number of sequences required in the alignment has fallen substantially. There is also substantial improvement in the accuracy of template-based models. Other areas-model refinement, accuracy estimation, and the structure of protein assemblies-have again yielded interesting results. CASP13 placed increased emphasis on the use of sparse data together with modeling and chemical crosslinking, SAXS, and NMR all yielded more mature results. This paper summarizes the key outcomes of CASP13. The special issue of PROTEINS contains papers describing the CASP13 assessments in each modeling category and contributions from the participants.


Assuntos
Sequência de Aminoácidos/genética , Biologia Computacional , Conformação Proteica , Proteínas/ultraestrutura , Humanos , Modelos Moleculares , Proteínas/química , Proteínas/genética , Espalhamento a Baixo Ângulo , Difração de Raios X
15.
Proteins ; 87(12): 1298-1314, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31589784

RESUMO

Small angle X-ray scattering (SAXS) measures comprehensive distance information on a protein's structure, which can constrain and guide computational structure prediction algorithms. Here, we evaluate structure predictions of 11 monomeric and oligomeric proteins for which SAXS data were collected and provided to predictors in the 13th round of the Critical Assessment of protein Structure Prediction (CASP13). The category for SAXS-assisted predictions made gains in certain areas for CASP13 compared to CASP12. Improvements included higher quality data with size exclusion chromatography-SAXS (SEC-SAXS) and better selection of targets and communication of results by CASP organizers. In several cases, we can track improvements in model accuracy with use of SAXS data. For hard multimeric targets where regular folding algorithms were unsuccessful, SAXS data helped predictors to build models better resembling the global shape of the target. For most models, however, no significant improvement in model accuracy at the domain level was registered from use of SAXS data, when rigorously comparing SAXS-assisted models to the best regular server predictions. To promote future progress in this category, we identify successes, challenges, and opportunities for improved strategies in prediction, assessment, and communication of SAXS data to predictors. An important observation is that, for many targets, SAXS data were inconsistent with crystal structures, suggesting that these proteins adopt different conformation(s) in solution. This CASP13 result, if representative of PDB structures and future CASP targets, may have substantive implications for the structure training databases used for machine learning, CASP, and use of prediction models for biology.


Assuntos
Biologia Computacional , Conformação Proteica , Proteínas/ultraestrutura , Algoritmos , Modelos Moleculares , Dobramento de Proteína , Proteínas/química , Proteínas/genética , Espalhamento a Baixo Ângulo , Soluções/química , Difração de Raios X
16.
Proteins ; 87(12): 1315-1332, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31603581

RESUMO

CASP13 has investigated the impact of sparse NMR data on the accuracy of protein structure prediction. NOESY and 15 N-1 H residual dipolar coupling data, typical of that obtained for 15 N,13 C-enriched, perdeuterated proteins up to about 40 kDa, were simulated for 11 CASP13 targets ranging in size from 80 to 326 residues. For several targets, two prediction groups generated models that are more accurate than those produced using baseline methods. Real NMR data collected for a de novo designed protein were also provided to predictors, including one data set in which only backbone resonance assignments were available. Some NMR-assisted prediction groups also did very well with these data. CASP13 also assessed whether incorporation of sparse NMR data improves the accuracy of protein structure prediction relative to nonassisted regular methods. In most cases, incorporation of sparse, noisy NMR data results in models with higher accuracy. The best NMR-assisted models were also compared with the best regular predictions of any CASP13 group for the same target. For six of 13 targets, the most accurate model provided by any NMR-assisted prediction group was more accurate than the most accurate model provided by any regular prediction group; however, for the remaining seven targets, one or more regular prediction method provided a more accurate model than even the best NMR-assisted model. These results suggest a novel approach for protein structure determination, in which advanced prediction methods are first used to generate structural models, and sparse NMR data is then used to validate and/or refine these models.


Assuntos
Espectroscopia de Ressonância Magnética/métodos , Modelos Moleculares , Conformação Proteica , Dobramento de Proteína , Proteínas/química , Algoritmos , Simulação por Computador , Cristalografia por Raios X , Reprodutibilidade dos Testes
18.
Proteins ; 86 Suppl 1: 202-214, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29314274

RESUMO

Experimental data offers empowering constraints for structure prediction. These constraints can be used to filter equivalently scored models or more powerfully within optimization functions toward prediction. In CASP12, Small Angle X-ray Scattering (SAXS) and Cross-Linking Mass Spectrometry (CLMS) data, measured on an exemplary set of novel fold targets, were provided to the CASP community of protein structure predictors. As solution-based techniques, SAXS and CLMS can efficiently measure states of the full-length sequence in its native solution conformation and assembly. However, this experimental data did not substantially improve prediction accuracy judged by fits to crystallographic models. One issue, beyond intrinsic limitations of the algorithms, was a disconnect between crystal structures and solution-based measurements. Our analyses show that many targets had substantial percentages of disordered regions (up to 40%) or were multimeric or both. Thus, solution measurements of flexibility and assembly support variations that may confound prediction algorithms trained on crystallographic data and expecting globular fully-folded monomeric proteins. Here, we consider the CLMS and SAXS data collected, the information in these solution measurements, and the challenges in incorporating them into computational prediction. As improvement opportunities were only partly realized in CASP12, we provide guidance on how data from the full-length biological unit and the solution state can better aid prediction of the folded monomer or subunit. We furthermore describe strategic integrations of solution measurements with computational prediction programs with the aim of substantially improving foundational knowledge and the accuracy of computational algorithms for biologically-relevant structure predictions for proteins in solution.


Assuntos
Biologia Computacional/métodos , Reagentes de Ligações Cruzadas/química , Espectrometria de Massas/métodos , Modelos Moleculares , Conformação Proteica , Proteínas/química , Espalhamento a Baixo Ângulo , Algoritmos , Humanos , Dobramento de Proteína , Difração de Raios X
19.
Proteins ; 86 Suppl 1: 345-360, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28833563

RESUMO

The record high 42 model accuracy estimation methods were tested in CASP12. The paper presents results of the assessment of these methods in the whole-model and per-residue accuracy modes. Scores from four different model evaluation packages were used as the "ground truth" for assessing accuracy of methods' estimates. They include a rigid-body score-GDT_TS, and three local-structure based scores-LDDT, CAD and SphereGrinder. The ability of methods to identify best models from among several available, predict model's absolute accuracy score, distinguish between good and bad models, predict accuracy of the coordinate error self-estimates, and discriminate between reliable and unreliable regions in the models was assessed. Single-model methods advanced to the point where they are better than clustering methods in picking the best models from decoy sets. On the other hand, consensus methods, taking advantage of the availability of large number of models for the same target protein, are still better in distinguishing between good and bad models and predicting local accuracy of models. The best accuracy estimation methods were shown to perform better with respect to the frozen in time reference clustering method and the results of the best method in the corresponding class of methods from the previous CASP. Top performing single-model methods were shown to do better than all but three CASP12 tertiary structure predictors when evaluated as model selectors.


Assuntos
Biologia Computacional/métodos , Modelos Moleculares , Conformação Proteica , Proteínas/química , Análise por Conglomerados , Bases de Dados de Proteínas , Humanos , Alinhamento de Sequência , Análise de Sequência de Proteína
20.
Proteins ; 86 Suppl 1: 321-334, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29159950

RESUMO

The article describes results of numerical evaluation of CASP12 models submitted on targets for which structural templates could be identified and for which servers produced models of relatively high accuracy. The emphasis is on analysis of details of models, and how well the models compete with experimental structures. Performance of contributing research groups is measured in terms of backbone accuracy, all-atom local geometry, and the ability to estimate local errors in models. Separate analyses for all participating groups and automatic servers were carried out. Compared with the last CASP, two years ago, there have been significant improvements in a number of areas, particularly the accuracy of protein backbone atoms, accuracy of sequence alignment between models and available structures, increased accuracy over that which can be obtained from simple copying of a closest template, and accuracy of modeling of sub-structures not present in the closest template. These advancements are likely associated with more effective strategies to build non-template regions of the targets ab initio, better algorithms to combine information from multiple templates, enhanced refinement methods, and better methods for estimating model accuracy.


Assuntos
Biologia Computacional/métodos , Modelos Moleculares , Modelos Estatísticos , Conformação Proteica , Proteínas/química , Bases de Dados de Proteínas , Humanos , Alinhamento de Sequência , Análise de Sequência de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA