1
|
Rosignoli S, Pacelli M, Manganiello F, Paiardini A. An outlook on structural biology after AlphaFold: tools, limits and perspectives. FEBS Open Bio 2025; 15:202-222. [PMID: 39313455 PMCID: PMC11788754 DOI: 10.1002/2211-5463.13902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/19/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
AlphaFold and similar groundbreaking, AI-based tools, have revolutionized the field of structural bioinformatics, with their remarkable accuracy in ab-initio protein structure prediction. This success has catalyzed the development of new software and pipelines aimed at incorporating AlphaFold's predictions, often focusing on addressing the algorithm's remaining challenges. Here, we present the current landscape of structural bioinformatics shaped by AlphaFold, and discuss how the field is dynamically responding to this revolution, with new software, methods, and pipelines. While the excitement around AI-based tools led to their widespread application, it is essential to acknowledge that their practical success hinges on their integration into established protocols within structural bioinformatics, often neglected in the context of AI-driven advancements. Indeed, user-driven intervention is still as pivotal in the structure prediction process as in complementing state-of-the-art algorithms with functional and biological knowledge.
Collapse
Affiliation(s)
- Serena Rosignoli
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| | - Maddalena Pacelli
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| | - Francesca Manganiello
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| | - Alessandro Paiardini
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| |
Collapse
|
2
|
Siciliano AJ, Zhao C, Liu T, Wang Z. EGG: Accuracy Estimation of Individual Multimeric Protein Models Using Deep Energy-Based Models and Graph Neural Networks. Int J Mol Sci 2024; 25:6250. [PMID: 38892437 PMCID: PMC11173161 DOI: 10.3390/ijms25116250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Reliable and accurate methods of estimating the accuracy of predicted protein models are vital to understanding their respective utility. Discerning how the quaternary structure conforms can significantly improve our collective understanding of cell biology, systems biology, disease formation, and disease treatment. Accurately determining the quality of multimeric protein models is still computationally challenging, as the space of possible conformations is significantly larger when proteins form in complex with one another. Here, we present EGG (energy and graph-based architectures) to assess the accuracy of predicted multimeric protein models. We implemented message-passing and transformer layers to infer the overall fold and interface accuracy scores of predicted multimeric protein models. When evaluated with CASP15 targets, our methods achieved promising results against single model predictors: fourth and third place for determining the highest-quality model when estimating overall fold accuracy and overall interface accuracy, respectively, and first place for determining the top three highest quality models when estimating both overall fold accuracy and overall interface accuracy.
Collapse
Affiliation(s)
- Andrew Jordan Siciliano
- Department of Computer Science, University of Miami, 1365 Memorial Drive, Coral Gables, FL 33124, USA; (A.J.S.); (T.L.)
| | - Chenguang Zhao
- Computer Information Sciences Department, St. Ambrose University, 518 W. Locust Street, Davenport, IA 52803, USA;
| | - Tong Liu
- Department of Computer Science, University of Miami, 1365 Memorial Drive, Coral Gables, FL 33124, USA; (A.J.S.); (T.L.)
| | - Zheng Wang
- Department of Computer Science, University of Miami, 1365 Memorial Drive, Coral Gables, FL 33124, USA; (A.J.S.); (T.L.)
| |
Collapse
|
3
|
Tasleem M, Hussein WM, El-Sayed AAAA, Alrehaily A. An In Silico Bioremediation Study to Identify Essential Residues of Metallothionein Enhancing the Bioaccumulation of Heavy Metals in Pseudomonas aeruginosa. Microorganisms 2023; 11:2262. [PMID: 37764106 PMCID: PMC10537150 DOI: 10.3390/microorganisms11092262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Microorganisms are ubiquitously present in the environment and exert significant influence on numerous natural phenomena. The soil and groundwater systems, precipitation, and effluent outfalls from factories, refineries, and waste treatment facilities are all sources of heavy metal contamination. For example, Madinah, Saudi Arabia, has alarmingly high levels of lead and cadmium. The non-essential minerals cadmium (Cd) and lead (Pb) have been linked to damage to vital organs. Bioremediation is an essential component in the process of cleaning up polluted soil and water where biological agents such as bacteria are used to remove the contaminants. It is demonstrated that Pseudomonas aeruginosa (P. aeruginosa) isolated from activated sludge was able to remove Cd and Pb from water. The protein sequence of metallothionein from P. aeruginosa was retrieved to explore it for physicoparameters, orthologs, domain, family, motifs, and conserved residues. The homology structure was generated, and models were validated. Docking of the best model with the heavy metals was carried out to inspect the intramolecular interactions. The target protein was found to belong to the "metallothionein_pro" family, containing six motifs, and showed a close orthologous relationship with other heavy metal-resistant bacteria. The best model was generated by Phyre2. In this study, three key residues of metallothionein were identified that participate in heavy metal (Pb and Cd) binding, viz., Ala33, Ser34, and Glu59. In addition, the study provides an essential basis to explore protein engineering for the optimum use of metallothionein protein to reduce/remove heavy metals from the environment.
Collapse
Affiliation(s)
- Munazzah Tasleem
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China;
| | - Wesam M. Hussein
- Chemistry Department, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia;
| | | | - Abdulwahed Alrehaily
- Biology Department, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia;
| |
Collapse
|
4
|
Wang J, Wang W, Shang Y. Protein Loop Modeling Using AlphaFold2. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:3306-3313. [PMID: 37037235 DOI: 10.1109/tcbb.2023.3264899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The functions of proteins are largely determined by their three-dimensional (3D) structures. Loop modeling tries to predict the conformation of a relatively short stretch of protein backbone and sidechain. It is a difficult problem due to conformational variability. Recently, AlphaFold2 has achieved outstanding results in 3-D protein structure prediction and is expected to perform well on loop modeling. In this paper, we investigate the performances of AlphaFold2 variants on popular loop modeling benchmark datasets and propose an efficient protocol of using AlphaFold2 for loop modeling, called IAFLoop. To predict the structure of a loop region, IAFLoop gives a moderately extended segment of the target loop region as input to AlphaFold2, runs a fast version of AlphaFold2 using a reduced database without ensembling, and uses RMSD based consensus scores to select the final output models. Our experimental results on benchmark datasets show that IAFLoop generated highly accurate loop models. It achieves comparable performance to the original application of AlphaFold2 in terms of RMSD error, and achieving much better results on some targets, while only using half of the time. Compared to the best previous methods, IAFLoop reduces the RMSD error by almost half on the 8-residual loop dataset, and more than 70% on the 12-residual loop dataset.
Collapse
|
5
|
Tatar G, Taskin Tok T, Ozpolat B, Ay M. Structure prediction of eukaryotic elongation factor-2 kinase and identification of the binding mechanisms of its inhibitors: homology modeling, molecular docking, and molecular dynamics simulation. J Biomol Struct Dyn 2022; 40:13355-13365. [PMID: 30880628 DOI: 10.1080/07391102.2019.1592024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Protein kinases emerged as one of the most successful families of drug targets due to their increased activity and involvement in mediating critical signal transduction pathways in cancer cells. Recent evidence suggests that eukaryotic elongation factor 2 kinase (eEF-2K) is a potential therapeutic target for treating some highly aggressive solid cancers, including lung, pancreatic and triple-negative breast cancers. Thus, several compounds have been developed for the inhibition of the enzyme activity, but they are not sufficiently specific and potent. Besides, the crystal structure of this kinase remains unknown. Hence, the functional organization and regulation of eEF-2K remain poorly characterized. For this purpose, we constructed a homology model of eEF-2K and then used docking methodology to better understanding the binding mechanism of eEF-2K with 58 compounds that have been proposed as existing inhibitors. The results of this analysis were compared with the experimental results and the compounds effective against eEF-2K were determined against eEF-2K as a result of both studies. And finally, molecular dynamics (MD) simulations were performed for the stability of eEF-2K with these compounds. According to these study defined that the binding mechanism of eEF-2K with inhibitors at the molecular level and elucidated the residues of eEF-2K that play an important role in enzyme selectivity and ligand affinity. This information may lead to new selective and potential drug molecules to be for inhibition of eEF-2K.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gizem Tatar
- Department of Bioinformatics and Computational Biology, Institute of Health Sciences, Gaziantep University, Gaziantep, Turkey
| | - Tugba Taskin Tok
- Department of Bioinformatics and Computational Biology, Institute of Health Sciences, Gaziantep University, Gaziantep, Turkey.,Department of Chemistry, Faculty of Arts and Sciences, Gaziantep University, Gaziantep, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas-Houston MD Anderson Cancer Center, Houston, USA
| | - Mehmet Ay
- Natural Products and Drug Research Laboratory, Department of Chemistry, Faculty of Science and Arts, Çanakkale Onsekiz Mart University Çanakkale, TURKEY
| |
Collapse
|
6
|
Stevens AO, He Y. Benchmarking the Accuracy of AlphaFold 2 in Loop Structure Prediction. Biomolecules 2022; 12:985. [PMID: 35883541 PMCID: PMC9312937 DOI: 10.3390/biom12070985] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 01/22/2023] Open
Abstract
The inhibition of protein-protein interactions is a growing strategy in drug development. In addition to structured regions, many protein loop regions are involved in protein-protein interactions and thus have been identified as potential drug targets. To effectively target such regions, protein structure is critical. Loop structure prediction is a challenging subgroup in the field of protein structure prediction because of the reduced level of conservation in protein sequences compared to the secondary structure elements. AlphaFold 2 has been suggested to be one of the greatest achievements in the field of protein structure prediction. The AlphaFold 2 predicted protein structures near the X-ray resolution in the Critical Assessment of protein Structure Prediction (CASP 14) competition in 2020. The purpose of this work is to survey the performance of AlphaFold 2 in specifically predicting protein loop regions. We have constructed an independent dataset of 31,650 loop regions from 2613 proteins (deposited after the AlphaFold 2 was trained) with both experimentally determined structures and AlphaFold 2 predicted structures. With extensive evaluation using our dataset, the results indicate that AlphaFold 2 is a good predictor of the structure of loop regions, especially for short loop regions. Loops less than 10 residues in length have an average Root Mean Square Deviation (RMSD) of 0.33 Å and an average the Template Modeling score (TM-score) of 0.82. However, we see that as the number of residues in a given loop increases, the accuracy of AlphaFold 2's prediction decreases. Loops more than 20 residues in length have an average RMSD of 2.04 Å and an average TM-score of 0.55. Such a correlation between accuracy and length of the loop is directly linked to the increase in flexibility. Moreover, AlphaFold 2 does slightly over-predict α-helices and β-strands in proteins.
Collapse
Affiliation(s)
- Amy O. Stevens
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Yi He
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131, USA;
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
7
|
Gromiha MM, Orengo CA, Sowdhamini R, Thornton AJM. Srinivasan (1962-2021) in Bioinformatics and beyond. Bioinformatics 2022; 38:2377-2379. [PMID: 35134112 PMCID: PMC9004639 DOI: 10.1093/bioinformatics/btac054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Affiliation(s)
- M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Christine A Orengo
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences (NCBS-TIFR), GKVK Campus, Bangalore, Karnataka 560065, India,Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India,Institute of Bioinformatics and Applied Biotechnology, Bangalore 560100, India
| | | |
Collapse
|
8
|
Affiliation(s)
- Frank Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore.,Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore.,School of Biological Sciences, Nanyang Technological University (NTU), Singapore
| | - Chandra Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore.,School of Biological Sciences, Nanyang Technological University (NTU), Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - Tom Blundell
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Wang W, Wang J, Li Z, Xu D, Shang Y. MUfoldQA_G: High-accuracy protein model QA via retraining and transformation. Comput Struct Biotechnol J 2021; 19:6282-6290. [PMID: 34900138 PMCID: PMC8636996 DOI: 10.1016/j.csbj.2021.11.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/10/2021] [Accepted: 11/14/2021] [Indexed: 11/21/2022] Open
Abstract
Protein tertiary structure prediction is an active research area and has attracted significant attention recently due to the success of AlphaFold from DeepMind. Methods capable of accurately evaluating the quality of predicted models are of great importance. In the past, although many model quality assessment (QA) methods have been developed, their accuracies are not consistently high across different QA performance metrics for diverse target proteins. In this paper, we propose MUfoldQA_G, a new multi-model QA method that aims at simultaneously optimizing Pearson correlation and average GDT-TS difference, two commonly used QA performance metrics. This method is based on two new algorithms MUfoldQA_Gp and MUfoldQA_Gr. MUfoldQA_Gp uses a new technique to combine information from protein templates and reference protein models to maximize the Pearson correlation QA metric. MUfoldQA_Gr employs a new machine learning technique that resamples training data and retrains adaptively to learn a consensus model that is better than naïve consensus while minimizing average GDT-TS difference. MUfoldQA_G uses a new method to combine the results of MUfoldQA_Gr and MUfoldQA_Gp so that the final QA prediction results achieve low average GDT-TS difference that is close to the results from MUfoldQA_Gr, while maintaining high Pearson correlation that is the same as the results from MUfoldQA_Gp. In CASP14 QA categories, MUfoldQA_G ranked No. 1 in Pearson correlation and No. 2 in average GDT-TS difference.
Collapse
Affiliation(s)
- Wenbo Wang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Junlin Wang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Zhaoyu Li
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Yi Shang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
10
|
The Methionine 549 and Leucine 552 Residues of Friedelin Synthase from Maytenus ilicifolia Are Important for Substrate Binding Specificity. Molecules 2021; 26:molecules26226806. [PMID: 34833897 PMCID: PMC8617677 DOI: 10.3390/molecules26226806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022] Open
Abstract
Friedelin, a pentacyclic triterpene found in the leaves of the Celastraceae species, demonstrates numerous biological activities and is a precursor of quinonemethide triterpenes, which are promising antitumoral agents. Friedelin is biosynthesized from the cyclization of 2,3-oxidosqualene, involving a series of rearrangements to form a ketone by deprotonation of the hydroxylated intermediate, without the aid of an oxidoreductase enzyme. Mutagenesis studies among oxidosqualene cyclases (OSCs) have demonstrated the influence of amino acid residues on rearrangements during substrate cyclization: loss of catalytic activity, stabilization, rearrangement control or specificity changing. In the present study, friedelin synthase from Maytenus ilicifolia (Celastraceae) was expressed heterologously in Saccharomyces cerevisiae. Site-directed mutagenesis studies were performed by replacing phenylalanine with tryptophan at position 473 (Phe473Trp), methionine with serine at position 549 (Met549Ser) and leucine with phenylalanine at position 552 (Leu552Phe). Mutation Phe473Trp led to a total loss of function; mutants Met549Ser and Leu552Phe interfered with the enzyme specificity leading to enhanced friedelin production, in addition to α-amyrin and β-amyrin. Hence, these data showed that methionine 549 and leucine 552 are important residues for the function of this synthase.
Collapse
|
11
|
Khan MKA, Akhtar S. Novel drug design and bioinformatics: an introduction. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2018-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
In the current era of high-throughput technology, where enormous amounts of biological data are generated day by day via various sequencing projects, thereby the staggering volume of biological targets deciphered. The discovery of new chemical entities and bioisosteres of relatively low molecular weight has been gaining high momentum in the pharmacopoeia, and traditional combinatorial design wherein chemical structure is used as an initial template for enhancing efficacy pharmacokinetic selectivity properties. Once the compound is identified, it undergoes ADMET filtration to ensure whether it has toxic and mutagenic properties or not. If the compound has no toxicity and mutagenicity is either considered a potential lead molecule. Understanding the mechanism of lead molecules with various biological targets is imperative to advance related functions for drug discovery and development. Notwithstanding, a tedious and costly process, taking around 10–15 years and costing around $4 billion, cascaded approached of Bioinformatics and Computational biology viz., structure-based drug design (SBDD) and cognate ligand-based drug design (LBDD) respectively rely on the availability of 3D structure of target biomacromolecules and vice versa has made this process easy and approachable. SBDD encompasses homology modelling, ligand docking, fragment-based drug design and molecular dynamics, while LBDD deals with pharmacophore mapping, QSAR, and similarity search. All the computational methods discussed herein, whether for target identification or novel ligand discovery, continuously evolve and facilitate cost-effective and reliable outcomes in an era of overwhelming data.
Collapse
Affiliation(s)
- Mohammad Kalim Ahmad Khan
- Department of Bioengineering, Faculty of Engineering , Integral University , Lucknow , Uttar Pradesh , 226026 , India
| | - Salman Akhtar
- Department of Bioengineering, Faculty of Engineering , Integral University , Lucknow , Uttar Pradesh , 226026 , India
| |
Collapse
|
12
|
Banerjee R, Sheet T, Banerjee S, Biondi B, Formaggio F, Toniolo C, Peggion C. C α-Methyl-l-valine: A Preferential Choice over α-Aminoisobutyric Acid for Designing Right-Handed α-Helical Scaffolds. Biochemistry 2021; 60:2704-2714. [PMID: 34463474 DOI: 10.1021/acs.biochem.1c00340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In synthetic peptides containing Gly and coded α-amino acids, one of the most common practices to enhance their helical extent is to incorporate a large number of l-Ala residues along with noncoded, strongly foldameric α-aminoisobutyric acid (Aib) units. Earlier studies have established that Aib-based peptides, with propensity for both the 310- and α-helices, have a tendency to form ordered three-dimensional structure that is much stronger than that exhibited by their l-Ala rich counterparts. However, the achiral nature of Aib induces an inherent, equal preference for the right- and left-handed helical conformations as found in Aib homopeptide stretches. This property poses challenges in the analysis of a model peptide helical conformation based on chirospectroscopic techniques like electronic circular dichroism (ECD), a very important tool for assigning secondary structures. To overcome such ambiguity, we have synthesized and investigated a thermally stable 14-mer peptide in which each of the Aib residues of our previously designed and reported analogue ABGY (where B stands for Aib) is replaced by Cα-methyl-l-valine (L-AMV). Analysis of the results described here from complementary ECD and 1H nuclear magnetic resonance spectroscopic techniques in a variety of environments firmly establishes that the L-AMV-containing peptide exhibits a significantly stronger preference compared to that of its Aib parent in terms of conferring α-helical character. Furthermore, being a chiral α-amino acid, L-AMV shows an intrinsic, extremely strong bias for a quite specific (right-handed) screw sense. These findings emphasize the relevance of L-AMV as a more appropriate unit for the design of right-handed α-helical peptide models that may be utilized as conformationally constrained scaffolds.
Collapse
Affiliation(s)
| | | | | | - Barbara Biondi
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.,Institute of Biomolecular Chemistry, Padova Unit, CNR, 35131 Padova, Italy
| | - Fernando Formaggio
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.,Institute of Biomolecular Chemistry, Padova Unit, CNR, 35131 Padova, Italy
| | - Claudio Toniolo
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.,Institute of Biomolecular Chemistry, Padova Unit, CNR, 35131 Padova, Italy
| | - Cristina Peggion
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.,Institute of Biomolecular Chemistry, Padova Unit, CNR, 35131 Padova, Italy
| |
Collapse
|
13
|
Khan SU, Ahemad N, Chuah LH, Naidu R, Htar TT. G protein-coupled estrogen receptor-1: homology modeling approaches and application in screening new GPER-1 modulators. J Biomol Struct Dyn 2020; 40:3325-3335. [PMID: 33164654 DOI: 10.1080/07391102.2020.1844059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
G protein-coupled receptors (GPCRs) belong to the largest family of protein targets comprising over 800 members in which at least 500 members are the therapeutic targets. Among the GPCRs, G protein-coupled estrogen receptor-1 (GPER-1) has shown to have the ability in estrogen signaling. As GPER-1 plays a critical role in several physiological responses, GPER-1 has been considered as a potential therapeutic target to treat estrogen-based cancers and other non-communicable diseases. However, the progress in the understanding of GPER-1 structure and function is relatively slow due to the availability of a only a few selective GPER-1 modulators. As with many GPCRs, the X-ray crystal structure of GPER-1 is yet to be resolved and thus has led the researchers to search for new GPER-1 modulators using homology models of GPER-1. In this review, we aim to summarize various approaches used in the generation of GPER-1 homology model and their applications that have resulted in new GPER-1 ligands.
Collapse
Affiliation(s)
- Shafi Ullah Khan
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia.,Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia.,Advanced Engineering Platform, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Thet Thet Htar
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
14
|
Santhoshkumar R, Yusuf A. In silico structural modeling and analysis of physicochemical properties of curcumin synthase (CURS1, CURS2, and CURS3) proteins of Curcuma longa. J Genet Eng Biotechnol 2020; 18:24. [PMID: 32617758 PMCID: PMC7332660 DOI: 10.1186/s43141-020-00041-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
Background Pharmaceutically important curcuminoid synthesis in C. longa is controlled by CURS1, CURS2, and CURS3 genes. The present study detected the physicochemical properties and structural characteristics including the secondary and 3D structure of CURS proteins. The primary, secondary, and tertiary structure of the CURS proteins were modeled and characterized using multiple bioinformatics tools such as ExPasy ProtParam tools, self-optimized prediction method with alignment (SOPMA), PSIPRED, and SWISS-MODEL. The predicted secondary structure of curcumin synthase provided an α-helix and random coil as the major components. The reliability of the modeled structure was confirmed using PROCHECK and QMEAN programs. Results The molecular weight of CURS1 is 21093.19 Da, theoretical pI as 4.93, and an aliphatic index of 99.19. Molecular weight of CURS2 and CURS3 proteins are 20266.13 Da and 20629.52 Da, theoretical pI as 5.28 and 4.96, and an aliphatic index of 89.30 and 86.37, respectively. In the predicted secondary structure of CURS proteins, alpha helices and random coils of CURS1, CUR2, and CURS3 were 42.72, 41.38, and 44.74% and 24.87, 31.03, and 17.89, respectively. The extended strands were 16.24, 19.40, and 17.89. QMEAN Z-score is − 0.83, − 0.89, and − 1.09 for CURS1, CURS2, and CURS3, respectively. Conclusion Prediction of the 3D model of a protein by in silico analysis is a highly challenging aspect to confirm the NMR or X-ray crystallographic data. This report can contribute to the understanding of the structure, physicochemical properties, structural motifs, and protein-protein interaction of CURS1, CUR2, and CURS3.
Collapse
Affiliation(s)
- R Santhoshkumar
- Interuniversity Centre for Plant Biotechnology, Department of Botany, University of Calicut, Malappuram, Kerala, 673635, India
| | - A Yusuf
- Interuniversity Centre for Plant Biotechnology, Department of Botany, University of Calicut, Malappuram, Kerala, 673635, India.
| |
Collapse
|
15
|
Wang W, Wang J, Xu D, Shang Y. Two New Heuristic Methods for Protein Model Quality Assessment. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1430-1439. [PMID: 30418914 PMCID: PMC8988942 DOI: 10.1109/tcbb.2018.2880202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Protein tertiary structure prediction is an important open challenge in bioinformatics and requires effective methods to accurately evaluate the quality of protein 3-D models generated computationally. Many quality assessment (QA) methods have been proposed over the past three decades. However, the accuracy or robustness is unsatisfactory for practical applications. In this paper, two new heuristic QA methods are proposed: MUfoldQA_S and MUfoldQA_C. The MUfoldQA_S is a quasi-single-model QA method that assesses the model quality based on the known protein structures with similar sequences. This algorithm can be directly applied to protein fragments without the necessity of building a full structural model. A BLOSUM-based heuristic is also introduced to help differentiate accurate templates from poor ones. In MUfoldQA_C, the ideas from MUfoldQA_S were combined with the consensus approach to create a multi-model QA method that could also utilize information from existing reference models and have demonstrated improved performance. Extensive experimental results of these two methods have shown significant improvement over existing methods. In addition, both methods have been blindly tested in the CASP12 world-wide competition in the protein structure prediction field and ranked as top performers in their respective categories.
Collapse
|
16
|
Wang W, Li Z, Wang J, Xu D, Shang Y. PSICA: a fast and accurate web service for protein model quality analysis. Nucleic Acids Res 2020; 47:W443-W450. [PMID: 31127307 PMCID: PMC6602450 DOI: 10.1093/nar/gkz402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/21/2019] [Accepted: 05/01/2019] [Indexed: 11/17/2022] Open
Abstract
This paper presents a new fast and accurate web service for protein model quality analysis, called PSICA (Protein Structural Information Conformity Analysis). It is designed to evaluate how much a tertiary model of a given protein primary sequence conforms to the known protein structures of similar protein sequences, and to evaluate the quality of predicted protein models. PSICA implements the MUfoldQA_S method, an efficient state-of-the-art protein model quality assessment (QA) method. In CASP12, MUfoldQA_S ranked No. 1 in the protein model QA select-20 category in terms of the difference between the predicted and true GDT-TS value of each model. For a given predicted 3D model, PSICA generates (i) predicted global GDT-TS value; (ii) interactive comparison between the model and other known protein structures; (iii) visualization of the predicted local quality of the model; and (iv) JSmol rendering of the model. Additionally, PSICA implements MUfoldQA_C, a new consensus method based on MUfoldQA_S. In CASP12, MUfoldQA_C ranked No. 1 in top 1 model GDT-TS loss on the select-20 QA category and No. 2 in the average difference between the predicted and true GDT-TS value of each model for both select-20 and best-150 QA categories. The PSICA server is freely available at http://qas.wangwb.com/∼wwr34/mufoldqa/index.html.
Collapse
Affiliation(s)
- Wenbo Wang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Zhaoyu Li
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Junlin Wang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Yi Shang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
17
|
Al Nasr K, Al-Haija QA. Forecasting the Growth of Structures from NMR and X-Ray Crystallography Experiments Released Per Year. JOURNAL OF INFORMATION & KNOWLEDGE MANAGEMENT 2020. [DOI: 10.1142/s0219649220400043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this paper, we present a forecasting scheme for the growth of molecular structures from NMR and X-ray Crystallography experimental techniques released every year by employing an autoregressive (AR) process. The proposed scheme maximises the forecasting accuracy by utilising the optimal AR process order. The optimal model order was derived as the model with the least prediction error. Therefore, the proposed scheme has been efficiently employed to model and predict the annual growth of structures-based NMR and X-ray Crystallography experimental data for the next decade 2019–2028 using the time series of the past 43 years of both experimental datasets. The experimental results showed that the optimal model order to estimate both datasets was [Formula: see text] which belongs to a forecasting accuracy of [Formula: see text], for both datasets. Indeed, such a high level of accuracy referred to the amount of linearity between the consecutive elements of the original times series. Hence, the forecasting results reveals of an exponential increasing behaviour in the future growth in the annual structures released from both NMR and X-ray Crystallography experiments.
Collapse
Affiliation(s)
- Kamal Al Nasr
- Department of Computer Science, Tennessee State University, Nashville, TN, USA
- University of Texas, San Antonio, TX, USA
| | - Qasem Abu Al-Haija
- Department of Computer and Information, Systems Engineering (CISE), Tennessee State University, Nashville, TN, USA
| |
Collapse
|
18
|
Shaker B, Yu MS, Lee J, Lee Y, Jung C, Na D. User guide for the discovery of potential drugs via protein structure prediction and ligand docking simulation. J Microbiol 2020; 58:235-244. [PMID: 32108318 DOI: 10.1007/s12275-020-9563-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 11/24/2022]
Abstract
Due to accumulating protein structure information and advances in computational methodologies, it has now become possible to predict protein-compound interactions. In biology, the classic strategy for drug discovery has been to manually screen multiple compounds (small scale) to identify potential drug compounds. Recent strategies have utilized computational drug discovery methods that involve predicting target protein structures, identifying active sites, and finding potential inhibitor compounds at large scale. In this protocol article, we introduce an in silico drug discovery protocol. Since multi-drug resistance of pathogenic bacteria remains a challenging problem to address, UDP-N-acetylmuramate-L-alanine ligase (murC) of Acinetobacter baumannii was used as an example, which causes nosocomial infection in hospital setups and is responsible for high mortality worldwide. This protocol should help microbiologists to expand their knowledge and research scope.
Collapse
Affiliation(s)
- Bilal Shaker
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Myung-Sang Yu
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jingyu Lee
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yongmin Lee
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Chanjin Jung
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dokyun Na
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
19
|
Nguyen SP, Li Z, Xu D, Shang Y. New Deep Learning Methods for Protein Loop Modeling. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:596-606. [PMID: 29990046 PMCID: PMC6580050 DOI: 10.1109/tcbb.2017.2784434] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Computational protein structure prediction is a long-standing challenge in bioinformatics. In the process of predicting protein 3D structures, it is common that parts of an experimental structure are missing or parts of a predicted structure need to be remodeled. The process of predicting local protein structures of particular regions is called loop modeling. In this paper, five new loop modeling methods based on machine learning techniques, called NearLooper, ConLooper, ResLooper, HyLooper1, and HyLooper2 are proposed. NearLooper is based on the nearest neighbor technique. ConLooper applies deep convolutional neural networks to predict ${\mathrm{C}}_{{{\alpha }}}$Cα atoms distance matrix as an orientation-independent representation of protein structure. ResLooper uses residual neural networks instead of deep convolutional neural networks. HyLooper1 combines the results of NearLooper and ConLooper while HyLooper2 combines NearLooper and ResLooper. Three commonly used benchmarks for loop modeling are used to compare the performance between these methods and existing state-of-the-art methods. The experiment results show promising performance in which our best method improves existing state-of-the-art methods by 28 and 54 percent of average RMSD on two datasets while being comparable on the other one.
Collapse
|
20
|
Golonko A, Pienkowski T, Swislocka R, Lazny R, Roszko M, Lewandowski W. Another look at phenolic compounds in cancer therapy the effect of polyphenols on ubiquitin-proteasome system. Eur J Med Chem 2019; 167:291-311. [PMID: 30776692 DOI: 10.1016/j.ejmech.2019.01.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/26/2022]
Abstract
Inhibitors of the ubiquitin-proteasome system (UPS) have been the object of research interests for many years because of their potential as anti-cancer agents. Research in this field is aimed at improving the specificity and safety of known proteasome inhibitors. Unfortunately, in vitro conditions do not reflect the processes taking place in the human body. Recent reports indicate that the components of human plasma affect the course of many signaling pathways, proteasome activity and the effectiveness of synthetic cytostatic drugs. Therefore, it is believed that the key issue is to determine the effects of components of the human diet, including effects of chemically active polyphenols on the ubiquitin-proteasome system activity in both physiological and pathological (cancerous) states. The following article summarizes the current knowledge on the direct and indirect synergistic and antagonistic effects between polyphenolic compounds present in the human diet and the efficiency of protein degradation via the UPS.
Collapse
Affiliation(s)
- Aleksandra Golonko
- Department of Food Analysis, Institute of Agricultural and Food Biotechnology, Rakowiecka 36, 02-532, Warsaw, Poland
| | - Tomasz Pienkowski
- Bialystok University of Technology, Faculty of Civil Engineering and Environmental Engineering, Department of Chemistry, Biology and Biotechnology, Wiejska 45E, 15-351, Bialystok, Poland
| | - Renata Swislocka
- Bialystok University of Technology, Faculty of Civil Engineering and Environmental Engineering, Department of Chemistry, Biology and Biotechnology, Wiejska 45E, 15-351, Bialystok, Poland
| | - Ryszard Lazny
- Institut of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245, Bialystok, Poland
| | - Marek Roszko
- Department of Food Analysis, Institute of Agricultural and Food Biotechnology, Rakowiecka 36, 02-532, Warsaw, Poland
| | - Wlodzimierz Lewandowski
- Department of Food Analysis, Institute of Agricultural and Food Biotechnology, Rakowiecka 36, 02-532, Warsaw, Poland.
| |
Collapse
|
21
|
Khare S, Bhasin M, Sahoo A, Varadarajan R. Protein model discrimination attempts using mutational sensitivity, predicted secondary structure, and model quality information. Proteins 2019; 87:326-336. [PMID: 30615225 DOI: 10.1002/prot.25654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/22/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023]
Abstract
Structure prediction methods often generate a large number of models for a target sequence. Even if the correct fold for the target sequence is sampled in this dataset, it is difficult to distinguish it from other decoy structures. An attempt to solve this problem using experimental mutational sensitivity data for the CcdB protein was described previously by exploiting the correlation of residue depth with mutational sensitivity (r ~ 0.6). We now show that such a correlation extends to four other proteins with localized active sites, and for which saturation mutagenesis datasets exist. We also examine whether incorporation of predicted secondary structure information and the DOPE model quality assessment score, in addition to mutational sensitivity, improves the accuracy of model discrimination using a decoy dataset of 163 targets from CASP. Although most CASP models would have been subjected to model quality assessment prior to submission, we find that the DOPE score makes a substantial contribution to the observed improvement. We therefore also applied the approach to CcdB and four other proteins for which reliable experimental mutational data exist and observe that inclusion of experimental mutational data results in a small qualitative improvement in model discrimination relative to that seen with just the DOPE score. This is largely because of our limited ability to quantitatively predict effects of point mutations on in vivo protein activity. Further improvements in the methodology are required to facilitate improved utilization of single mutant data.
Collapse
Affiliation(s)
- Shruti Khare
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Munmun Bhasin
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Anusmita Sahoo
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.,Chemical Biology Unit, Jawaharlal Nehru Center for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
22
|
Sadeghi S, Poorebrahim M, Rahimi H, Karimipoor M, Azadmanesh K, Khorramizadeh MR, Teimoori-Toolabi L. In silico studying of the whole protein structure and dynamics of Dickkopf family members showed that N-terminal domain of Dickkopf 2 in contrary to other Dickkopfs facilitates its interaction with low density lipoprotein receptor related protein 5/6. J Biomol Struct Dyn 2018; 37:2564-2580. [DOI: 10.1080/07391102.2018.1491891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Solmaz Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Hamzeh Rahimi
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Mohammad Reza Khorramizadeh
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Nithin C, Ghosh P, Bujnicki JM. Bioinformatics Tools and Benchmarks for Computational Docking and 3D Structure Prediction of RNA-Protein Complexes. Genes (Basel) 2018; 9:genes9090432. [PMID: 30149645 PMCID: PMC6162694 DOI: 10.3390/genes9090432] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/26/2018] [Accepted: 08/21/2018] [Indexed: 12/29/2022] Open
Abstract
RNA-protein (RNP) interactions play essential roles in many biological processes, such as regulation of co-transcriptional and post-transcriptional gene expression, RNA splicing, transport, storage and stabilization, as well as protein synthesis. An increasing number of RNP structures would aid in a better understanding of these processes. However, due to the technical difficulties associated with experimental determination of macromolecular structures by high-resolution methods, studies on RNP recognition and complex formation present significant challenges. As an alternative, computational prediction of RNP interactions can be carried out. Structural models obtained by theoretical predictive methods are, in general, less reliable compared to models based on experimental measurements but they can be sufficiently accurate to be used as a basis for to formulating functional hypotheses. In this article, we present an overview of computational methods for 3D structure prediction of RNP complexes. We discuss currently available methods for macromolecular docking and for scoring 3D structural models of RNP complexes in particular. Additionally, we also review benchmarks that have been developed to assess the accuracy of these methods.
Collapse
Affiliation(s)
- Chandran Nithin
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, ul. Ks. Trojdena 4, PL-02-109 Warsaw, Poland.
| | - Pritha Ghosh
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, ul. Ks. Trojdena 4, PL-02-109 Warsaw, Poland.
| | - Janusz M Bujnicki
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, ul. Ks. Trojdena 4, PL-02-109 Warsaw, Poland.
- Bioinformatics Laboratory, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, ul. Umultowska 89, PL-61-614 Poznan, Poland.
| |
Collapse
|
24
|
Tang Z, Jin W, Tang Y, Wang Y, Wang C, Zheng X, Sun W, Liu M, Zheng T, Chen H, Wu Q, Shan Z, Bu T, Li C. Research on homology modeling, molecular docking of the cellulase and highly expression of the key enzyme (Bgl) in Pichia pastoris. Int J Biol Macromol 2018; 115:1079-1087. [DOI: 10.1016/j.ijbiomac.2018.04.135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 10/17/2022]
|
25
|
Sahlgren C, Meinander A, Zhang H, Cheng F, Preis M, Xu C, Salminen TA, Toivola D, Abankwa D, Rosling A, Karaman DŞ, Salo-Ahen OMH, Österbacka R, Eriksson JE, Willför S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N. Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems. Adv Healthc Mater 2017; 6. [PMID: 28892296 DOI: 10.1002/adhm.201700258] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/04/2017] [Indexed: 12/13/2022]
Abstract
Approaches to increase the efficiency in developing drugs and diagnostics tools, including new drug delivery and diagnostic technologies, are needed for improved diagnosis and treatment of major diseases and health problems such as cancer, inflammatory diseases, chronic wounds, and antibiotic resistance. Development within several areas of research ranging from computational sciences, material sciences, bioengineering to biomedical sciences and bioimaging is needed to realize innovative drug development and diagnostic (DDD) approaches. Here, an overview of recent progresses within key areas that can provide customizable solutions to improve processes and the approaches taken within DDD is provided. Due to the broadness of the area, unfortunately all relevant aspects such as pharmacokinetics of bioactive molecules and delivery systems cannot be covered. Tailored approaches within (i) bioinformatics and computer-aided drug design, (ii) nanotechnology, (iii) novel materials and technologies for drug delivery and diagnostic systems, and (iv) disease models to predict safety and efficacy of medicines under development are focused on. Current developments and challenges ahead are discussed. The broad scope reflects the multidisciplinary nature of the field of DDD and aims to highlight the convergence of biological, pharmaceutical, and medical disciplines needed to meet the societal challenges of the 21st century.
Collapse
Affiliation(s)
- Cecilia Sahlgren
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Annika Meinander
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Hongbo Zhang
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Fang Cheng
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Maren Preis
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Chunlin Xu
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Tiina A. Salminen
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Diana Toivola
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Center for Disease Modeling; University of Turku; FI-20520 Turku Finland
| | - Daniel Abankwa
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Ari Rosling
- Faculty of Science and Engineering; Polymer Technologies; Åbo Akademi University; FI-20500 Turku Finland
| | - Didem Şen Karaman
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Outi M. H. Salo-Ahen
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Ronald Österbacka
- Faculty of Science and Engineering; Physics; Åbo Akademi University; FI-20500 Turku Finland
| | - John E. Eriksson
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
| | - Stefan Willför
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Ion Petre
- Faculty of Science and Engineering; Computer Science; Åbo Akademi University; FI-20500 Turku Finland
| | - Jouko Peltonen
- Faculty of Science and Engineering; Physical Chemistry; Åbo Akademi University; FI-20500 Turku Finland
| | - Reko Leino
- Faculty of Science and Engineering; Organic Chemistry; Johan Gadolin Process Chemistry Centre; Åbo Akademi University; FI-20500 Turku Finland
| | - Mark Johnson
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Jessica Rosenholm
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Niklas Sandler
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| |
Collapse
|
26
|
Sahlgren C, Meinander A, Zhang H, Cheng F, Preis M, Xu C, Salminen TA, Toivola D, Abankwa D, Rosling A, Karaman DŞ, Salo-Ahen OMH, Österbacka R, Eriksson JE, Willför S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N. Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems. Adv Healthc Mater 2017. [DOI: 10.1002/adhm.201700258 10.1002/adhm.201700258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Affiliation(s)
- Cecilia Sahlgren
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Annika Meinander
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Hongbo Zhang
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Fang Cheng
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Maren Preis
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Chunlin Xu
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Tiina A. Salminen
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Diana Toivola
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Center for Disease Modeling; University of Turku; FI-20520 Turku Finland
| | - Daniel Abankwa
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Ari Rosling
- Faculty of Science and Engineering; Polymer Technologies; Åbo Akademi University; FI-20500 Turku Finland
| | - Didem Şen Karaman
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Outi M. H. Salo-Ahen
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Ronald Österbacka
- Faculty of Science and Engineering; Physics; Åbo Akademi University; FI-20500 Turku Finland
| | - John E. Eriksson
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
| | - Stefan Willför
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Ion Petre
- Faculty of Science and Engineering; Computer Science; Åbo Akademi University; FI-20500 Turku Finland
| | - Jouko Peltonen
- Faculty of Science and Engineering; Physical Chemistry; Åbo Akademi University; FI-20500 Turku Finland
| | - Reko Leino
- Faculty of Science and Engineering; Organic Chemistry; Johan Gadolin Process Chemistry Centre; Åbo Akademi University; FI-20500 Turku Finland
| | - Mark Johnson
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Jessica Rosenholm
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Niklas Sandler
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| |
Collapse
|
27
|
Kaushik AC, Sahi S. Insights into unbound-bound states of GPR142 receptor in a membrane-aqueous system using molecular dynamics simulations. J Biomol Struct Dyn 2017; 36:1788-1805. [PMID: 28571491 DOI: 10.1080/07391102.2017.1335234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
G protein coupled receptors (GPCRs) are source machinery in signal transduction pathways and being one of the major therapeutic targets play a significant in drug discovery. GPR142, an orphan GPCR, has been implicated in the regulation of insulin, thereby having a crucial role in Type II diabetes management. Deciphering of the structures of orphan, GPCRs (O-GPCRs) offer better prospects for advancements in research in ion translocation and transduction of extracellular signals. As the crystallographic structure of GPR142 is not available in PDB, therefore, threading and ab initio-based approaches were used for 3D modeling of GPR142. Molecular dynamic simulations (900 ns) were performed on the 3D model of GPR142 and complexes of GPR142 with top five hits, obtained through virtual screening, embedded in lipid bilayer with aqueous system using OPLS force field. Compound 1, 3, and 4 may act as scaffolds for designing potential lead agonists for GPR142. The finding of GPR142 MD simulation study provides more comprehensive representation of the functional properties. The concern for Type II diabetes is increasing worldwide and successful treatment of this disease demands novel drugs with better efficacy.
Collapse
Affiliation(s)
- Aman Chandra Kaushik
- a School of Biotechnology , Gautam Buddha University , Greater Noida , Uttar Pradesh , India
| | - Shakti Sahi
- a School of Biotechnology , Gautam Buddha University , Greater Noida , Uttar Pradesh , India
| |
Collapse
|
28
|
Souza-Moreira TM, Alves TB, Pinheiro KA, Felippe LG, De Lima GMA, Watanabe TF, Barbosa CC, Santos VAFFM, Lopes NP, Valentini SR, Guido RVC, Furlan M, Zanelli CF. Friedelin Synthase from Maytenus ilicifolia: Leucine 482 Plays an Essential Role in the Production of the Most Rearranged Pentacyclic Triterpene. Sci Rep 2016; 6:36858. [PMID: 27874020 PMCID: PMC5118845 DOI: 10.1038/srep36858] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/20/2016] [Indexed: 11/26/2022] Open
Abstract
Among the biologically active triterpenes, friedelin has the most-rearranged structure produced by the oxidosqualene cyclases and is the only one containing a cetonic group. In this study, we cloned and functionally characterized friedelin synthase and one cycloartenol synthase from Maytenus ilicifolia (Celastraceae). The complete coding sequences of these 2 genes were cloned from leaf mRNA, and their functions were characterized by heterologous expression in yeast. The cycloartenol synthase sequence is very similar to other known OSCs of this type (approximately 80% identity), although the M. ilicifolia friedelin synthase amino acid sequence is more related to β-amyrin synthases (65-74% identity), which is similar to the friedelin synthase cloned from Kalanchoe daigremontiana. Multiple sequence alignments demonstrated the presence of a leucine residue two positions upstream of the friedelin synthase Asp-Cys-Thr-Ala-Glu (DCTAE) active site motif, while the vast majority of OSCs identified so far have a valine or isoleucine residue at the same position. The substitution of the leucine residue with valine, threonine or isoleucine in M. ilicifolia friedelin synthase interfered with substrate recognition and lead to the production of different pentacyclic triterpenes. Hence, our data indicate a key role for the leucine residue in the structure and function of this oxidosqualene cyclase.
Collapse
Affiliation(s)
- Tatiana M. Souza-Moreira
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Thaís B. Alves
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Karina A. Pinheiro
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Lidiane G. Felippe
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Gustavo M. A. De Lima
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP 13563-120, Brazil
| | - Tatiana F. Watanabe
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Cristina C. Barbosa
- Faculdade de Ciências Farmacêuticas, Univ. Estadual Paulista-UNESP, Rod. Araraquara-Jaú km 1, Araraquara, SP 14801-902, Brazil
| | - Vânia A. F. F. M. Santos
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Norberto P. Lopes
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Avenida do Café s/n, Monte Alegre, Ribeirão Preto, SP 14040-903, Brazil
| | - Sandro R. Valentini
- Faculdade de Ciências Farmacêuticas, Univ. Estadual Paulista-UNESP, Rod. Araraquara-Jaú km 1, Araraquara, SP 14801-902, Brazil
| | - Rafael V. C. Guido
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP 13563-120, Brazil
| | - Maysa Furlan
- Instituto de Química, Univ. Estadual Paulista-UNESP, Rua Prof. Francisco Degni, 55, Quitandinha, Araraquara, SP 14800-060, Brazil
| | - Cleslei F. Zanelli
- Faculdade de Ciências Farmacêuticas, Univ. Estadual Paulista-UNESP, Rod. Araraquara-Jaú km 1, Araraquara, SP 14801-902, Brazil
| |
Collapse
|
29
|
Santana JO, Freire L, de Sousa AO, Fontes Soares VL, Gramacho KP, Pirovani CP. Characterization of the legumains encoded by the genome of Theobroma cacao L. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2016; 98:162-170. [PMID: 26691061 DOI: 10.1016/j.plaphy.2015.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/29/2015] [Accepted: 11/16/2015] [Indexed: 06/05/2023]
Abstract
Legumains are cysteine proteases related to plant development, protein degradation, programmed cell death, and defense against pathogens. In this study, we have identified and characterized three legumains encoded by Theobroma cacao genome through in silico analyses, three-dimensional modeling, genetic expression pattern in different tissues and as a response to the inoculation of Moniliophthora perniciosa fungus. The three proteins were named TcLEG3, TcLEG6, and TcLEG9. Histidine and cysteine residue which are part of the catalytic site were conserved among the proteins, and they remained parallel in the loop region in the 3D modeling. Three-dimensional modeling showed that the propeptide, which is located in the terminal C region of legumains blocks the catalytic cleft. Comparing dendrogram data with the relative expression analysis, indicated that TcLEG3 is related to the seed legumain group, TcLEG6 is related with the group of embryogenesis activities, and protein TcLEG9, with processes regarding the vegetative group. Furthermore, the expression analyses proposes a significant role for the three legumains during the development of Theobroma cacao and in its interaction with M. perniciosa.
Collapse
Affiliation(s)
| | - Laís Freire
- Biotechnology and Genetics Center, State University of Santa Cruz, 45662-900 Ilhéus, BA, Brazil
| | | | | | | | - Carlos Priminho Pirovani
- Biotechnology and Genetics Center, State University of Santa Cruz, 45662-900 Ilhéus, BA, Brazil.
| |
Collapse
|
30
|
Borguesan B, e Silva MB, Grisci B, Inostroza-Ponta M, Dorn M. APL: An angle probability list to improve knowledge-based metaheuristics for the three-dimensional protein structure prediction. Comput Biol Chem 2015; 59 Pt A:142-57. [DOI: 10.1016/j.compbiolchem.2015.08.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 08/05/2015] [Accepted: 08/17/2015] [Indexed: 10/23/2022]
|
31
|
Shameer K, Tripathi LP, Kalari KR, Dudley JT, Sowdhamini R. Interpreting functional effects of coding variants: challenges in proteome-scale prediction, annotation and assessment. Brief Bioinform 2015; 17:841-62. [PMID: 26494363 DOI: 10.1093/bib/bbv084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Indexed: 12/20/2022] Open
Abstract
Accurate assessment of genetic variation in human DNA sequencing studies remains a nontrivial challenge in clinical genomics and genome informatics. Ascribing functional roles and/or clinical significances to single nucleotide variants identified from a next-generation sequencing study is an important step in genome interpretation. Experimental characterization of all the observed functional variants is yet impractical; thus, the prediction of functional and/or regulatory impacts of the various mutations using in silico approaches is an important step toward the identification of functionally significant or clinically actionable variants. The relationships between genotypes and the expressed phenotypes are multilayered and biologically complex; such relationships present numerous challenges and at the same time offer various opportunities for the design of in silico variant assessment strategies. Over the past decade, many bioinformatics algorithms have been developed to predict functional consequences of single nucleotide variants in the protein coding regions. In this review, we provide an overview of the bioinformatics resources for the prediction, annotation and visualization of coding single nucleotide variants. We discuss the currently available approaches and major challenges from the perspective of protein sequence, structure, function and interactions that require consideration when interpreting the impact of putatively functional variants. We also discuss the relevance of incorporating integrated workflows for predicting the biomedical impact of the functionally important variations encoded in a genome, exome or transcriptome. Finally, we propose a framework to classify variant assessment approaches and strategies for incorporation of variant assessment within electronic health records.
Collapse
|
32
|
Kumalo HM, Bhakat S, Soliman ME. Heat-shock protein 90 (Hsp90) as anticancer target for drug discovery: an ample computational perspective. Chem Biol Drug Des 2015; 86:1131-60. [PMID: 25958815 DOI: 10.1111/cbdd.12582] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There are over 100 different types of cancer, and each is classified based on the type of cell that is initially affected. If left untreated, cancer can result in serious health problems and eventually death. Recently, the paradigm of cancer chemotherapy has evolved to use a combination approach, which involves the use of multiple drugs each of which targets an individual protein. Inhibition of heat-shock protein 90 (Hsp90) is one of the novel key cancer targets. Because of its ability to target several signaling pathways, Hsp90 inhibition emerged as a useful strategy to treat a wide variety of cancers. Molecular modeling approaches and methodologies have become 'close counterparts' to experiments in drug design and discovery workflows. A wide range of molecular modeling approaches have been developed, each of which has different objectives and outcomes. In this review, we provide an up-to-date systematic overview on the different computational models implemented toward the design of Hsp90 inhibitors as anticancer agents. Although this is the main emphasis of this review, different topics such as background and current statistics of cancer, different anticancer targets including Hsp90, and the structure and function of Hsp90 from an experimental perspective, for example, X-ray and NMR, are also addressed in this report. To the best of our knowledge, this review is the first account, which comprehensively outlines various molecular modeling efforts directed toward identification of anticancer drugs targeting Hsp90. We believe that the information, methods, and perspectives highlighted in this report would assist researchers in the discovery of potential anticancer agents.
Collapse
Affiliation(s)
- Hezekiel M Kumalo
- School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, 4001, South Africa
| | - Soumendranath Bhakat
- School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, 4001, South Africa.,Division of Biophysical Chemistry, Lund University, P.O. Box 124, SE-22100, Lund, Sweden
| | - Mahmoud E Soliman
- School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, 4001, South Africa
| |
Collapse
|
33
|
Three-dimensional protein structure prediction: Methods and computational strategies. Comput Biol Chem 2014; 53PB:251-276. [DOI: 10.1016/j.compbiolchem.2014.10.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 01/01/2023]
|
34
|
|
35
|
Nguyen SP, Shang Y, Xu D. DL-PRO: A Novel Deep Learning Method for Protein Model Quality Assessment. PROCEEDINGS OF ... INTERNATIONAL JOINT CONFERENCE ON NEURAL NETWORKS. INTERNATIONAL JOINT CONFERENCE ON NEURAL NETWORKS 2014; 2014:2071-2078. [PMID: 25392745 PMCID: PMC4226404 DOI: 10.1109/ijcnn.2014.6889891] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Computational protein structure prediction is very important for many applications in bioinformatics. In the process of predicting protein structures, it is essential to accurately assess the quality of generated models. Although many single-model quality assessment (QA) methods have been developed, their accuracy is not high enough for most real applications. In this paper, a new approach based on C-α atoms distance matrix and machine learning methods is proposed for single-model QA and the identification of native-like models. Different from existing energy/scoring functions and consensus approaches, this new approach is purely geometry based. Furthermore, a novel algorithm based on deep learning techniques, called DL-Pro, is proposed. For a protein model, DL-Pro uses its distance matrix that contains pairwise distances between two residues' C-α atoms in the model, which sometimes is also called contact map, as an orientation-independent representation. From training examples of distance matrices corresponding to good and bad models, DL-Pro learns a stacked autoencoder network as a classifier. In experiments on selected targets from the Critical Assessment of Structure Prediction (CASP) competition, DL-Pro obtained promising results, outperforming state-of-the-art energy/scoring functions, including OPUS-CA, DOPE, DFIRE, and RW.
Collapse
Affiliation(s)
- Son P. Nguyen
- Department of Computer Science, University of Missouri, Columbia, MO 65211 USA
| | - Yi Shang
- Department of Computer Science, University of Missouri, Columbia, MO 65211 USA
| | - Dong Xu
- Department of Computer Science, University of Missouri, Columbia, MO 65211 USA. Christopher S. Bond Life Science Center, University of Missouri at Columbia
| |
Collapse
|
36
|
Chen Y, Shang Y, Xu D. Multi-Dimensional Scaling and MODELLER-Based Evolutionary Algorithms for Protein Model Refinement. PROCEEDINGS OF THE ... CONGRESS ON EVOLUTIONARY COMPUTATION. CONGRESS ON EVOLUTIONARY COMPUTATION 2014; 2014:1038-1045. [PMID: 25844403 PMCID: PMC4380876 DOI: 10.1109/cec.2014.6900443] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Protein structure prediction, i.e., computationally predicting the three-dimensional structure of a protein from its primary sequence, is one of the most important and challenging problems in bioinformatics. Model refinement is a key step in the prediction process, where improved structures are constructed based on a pool of initially generated models. Since the refinement category was added to the biennial Critical Assessment of Structure Prediction (CASP) in 2008, CASP results show that it is a challenge for existing model refinement methods to improve model quality consistently. This paper presents three evolutionary algorithms for protein model refinement, in which multidimensional scaling(MDS), the MODELLER software, and a hybrid of both are used as crossover operators, respectively. The MDS-based method takes a purely geometrical approach and generates a child model by combining the contact maps of multiple parents. The MODELLER-based method takes a statistical and energy minimization approach, and uses the remodeling module in MODELLER program to generate new models from multiple parents. The hybrid method first generates models using the MDS-based method and then run them through the MODELLER-based method, aiming at combining the strength of both. Promising results have been obtained in experiments using CASP datasets. The MDS-based method improved the best of a pool of predicted models in terms of the global distance test score (GDT-TS) in 9 out of 16test targets.
Collapse
Affiliation(s)
- Yan Chen
- Yan Chen, Yi Shang, and Dong Xu are with the Department of Computer Science, University of Missouri, Columbia, MO 65211 USA. Dong Xu is also with the Christopher S. Bond Life Science Center, University of Missouri. (, , and )
| | - Yi Shang
- Yan Chen, Yi Shang, and Dong Xu are with the Department of Computer Science, University of Missouri, Columbia, MO 65211 USA. Dong Xu is also with the Christopher S. Bond Life Science Center, University of Missouri. (, , and )
| | - Dong Xu
- Yan Chen, Yi Shang, and Dong Xu are with the Department of Computer Science, University of Missouri, Columbia, MO 65211 USA. Dong Xu is also with the Christopher S. Bond Life Science Center, University of Missouri. (, , and )
| |
Collapse
|
37
|
Abstract
Structural proteomics aims to understand the structural basis of protein interactions and functions. A prerequisite for this is the availability of 3D protein structures that mediate the biochemical interactions. The explosion in the number of available gene sequences set the stage for the next step in genome-scale projects -- to obtain 3D structures for each protein. To achieve this ambitious goal, the slow and costly structure determination experiments are supplemented with theoretical approaches. The current state and recent advances in structure modeling approaches are reviewed here, with special emphasis on comparative protein structure modeling techniques.
Collapse
Affiliation(s)
- András Fiser
- Department of Biochemistry, Seaver Foundation Center for Bioinformatics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| |
Collapse
|
38
|
Webb B, Eswar N, Fan H, Khuri N, Pieper U, Dong G, Sali A. Comparative Modeling of Drug Target Proteins☆. REFERENCE MODULE IN CHEMISTRY, MOLECULAR SCIENCES AND CHEMICAL ENGINEERING 2014. [PMCID: PMC7157477 DOI: 10.1016/b978-0-12-409547-2.11133-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this perspective, we begin by describing the comparative protein structure modeling technique and the accuracy of the corresponding models. We then discuss the significant role that comparative prediction plays in drug discovery. We focus on virtual ligand screening against comparative models and illustrate the state-of-the-art by a number of specific examples.
Collapse
|
39
|
Rajapandiyan K, Shanthi S, Vijayalakshmi P, Daisy P, Murugan M, Ranjitsingh AJA. Prevalence of extended-spectrum β-lactamase-producing multidrug-resistant Escherichia coli among isolates from community-acquired infections and in silico structural modeling of an ESBL protein. Microb Drug Resist 2013; 20:170-6. [PMID: 24228708 DOI: 10.1089/mdr.2013.0088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Escherichia coli is a common major cause of bacterial infections in tea tribe patients of the northeast region of Assam, India. In this study, we documented multidrug resistance (MDR) and the prevalence of extended-spectrum β-lactamases (ESBLs) among 148 E. coli strains that were isolated from bacterial infections in tea tribe patients who had a history of self-medication. High prevalence of resistance to ampicillin (82%), amoxicillin (68%), cefixime (60%), norfloxacin (60%), nalidixic acid (60%), and co-trimoxazole (53%) was observed. Of 148 E. coli isolates, 38 (26%) were confirmed as ESBL producers. The ESBL genes were sequenced from highly resistant ESBL producing E. coli isolates. Molecular modeling was performed using MODELLER 9v10 software to determine the three-dimensional structure of a protein. This result indicates that the prevailing reason for the high prevalence of antibiotic resistance in this community is prior exposure to low-quality antibiotics, hence MDR in E. coli is increasing. ESBLs are enzymes that are produced by resistant bacteria that hydrolyze advanced generations of cephalosporin antibiotics and cause resistance, even in patients with community-acquired infections. So our results provide a framework for understanding the structure and possible binding sites of ESBL proteins for drug targeting, and the results were found to be reliable.
Collapse
|
40
|
Honarparvar B, Govender T, Maguire GEM, Soliman MES, Kruger HG. Integrated Approach to Structure-Based Enzymatic Drug Design: Molecular Modeling, Spectroscopy, and Experimental Bioactivity. Chem Rev 2013; 114:493-537. [DOI: 10.1021/cr300314q] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Bahareh Honarparvar
- Catalysis
and Peptide Research Unit and ‡School of Health Sciences, University of KwaZulu Natal, Durban 4001, South Africa
| | - Thavendran Govender
- Catalysis
and Peptide Research Unit and ‡School of Health Sciences, University of KwaZulu Natal, Durban 4001, South Africa
| | - Glenn E. M. Maguire
- Catalysis
and Peptide Research Unit and ‡School of Health Sciences, University of KwaZulu Natal, Durban 4001, South Africa
| | - Mahmoud E. S. Soliman
- Catalysis
and Peptide Research Unit and ‡School of Health Sciences, University of KwaZulu Natal, Durban 4001, South Africa
| | - Hendrik G. Kruger
- Catalysis
and Peptide Research Unit and ‡School of Health Sciences, University of KwaZulu Natal, Durban 4001, South Africa
| |
Collapse
|
41
|
Ding W, Xie J, Dai D, Zhang H, Xie H, Zhang W. CNNcon: improved protein contact maps prediction using cascaded neural networks. PLoS One 2013; 8:e61533. [PMID: 23626696 PMCID: PMC3634008 DOI: 10.1371/journal.pone.0061533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 03/11/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUNDS Despite continuing progress in X-ray crystallography and high-field NMR spectroscopy for determination of three-dimensional protein structures, the number of unsolved and newly discovered sequences grows much faster than that of determined structures. Protein modeling methods can possibly bridge this huge sequence-structure gap with the development of computational science. A grand challenging problem is to predict three-dimensional protein structure from its primary structure (residues sequence) alone. However, predicting residue contact maps is a crucial and promising intermediate step towards final three-dimensional structure prediction. Better predictions of local and non-local contacts between residues can transform protein sequence alignment to structure alignment, which can finally improve template based three-dimensional protein structure predictors greatly. METHODS CNNcon, an improved multiple neural networks based contact map predictor using six sub-networks and one final cascade-network, was developed in this paper. Both the sub-networks and the final cascade-network were trained and tested with their corresponding data sets. While for testing, the target protein was first coded and then input to its corresponding sub-networks for prediction. After that, the intermediate results were input to the cascade-network to finish the final prediction. RESULTS The CNNcon can accurately predict 58.86% in average of contacts at a distance cutoff of 8 Å for proteins with lengths ranging from 51 to 450. The comparison results show that the present method performs better than the compared state-of-the-art predictors. Particularly, the prediction accuracy keeps steady with the increase of protein sequence length. It indicates that the CNNcon overcomes the thin density problem, with which other current predictors have trouble. This advantage makes the method valuable to the prediction of long length proteins. As a result, the effective prediction of long length proteins could be possible by the CNNcon.
Collapse
Affiliation(s)
- Wang Ding
- School of Computer Engineering and Science, Shanghai University, Shanghai, People’s Republic of China
| | - Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, People’s Republic of China
- Institute of Systems Biology, Shanghai University, Shanghai, People’s Republic of China
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
| | - Dongbo Dai
- School of Computer Engineering and Science, Shanghai University, Shanghai, People’s Republic of China
| | - Huiran Zhang
- School of Computer Engineering and Science, Shanghai University, Shanghai, People’s Republic of China
| | - Hao Xie
- College of Stomatology, Wuhan University, Wuhan, People’s Republic of China
| | - Wu Zhang
- School of Computer Engineering and Science, Shanghai University, Shanghai, People’s Republic of China
- Institute of Systems Biology, Shanghai University, Shanghai, People’s Republic of China
- * E-mail:
| |
Collapse
|
42
|
Vyas VK, Ukawala RD, Ghate M, Chintha C. Homology modeling a fast tool for drug discovery: current perspectives. Indian J Pharm Sci 2012. [PMID: 23204616 PMCID: PMC3507339 DOI: 10.4103/0250-474x.102537] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Major goal of structural biology involve formation of protein-ligand complexes; in which the protein molecules act energetically in the course of binding. Therefore, perceptive of protein-ligand interaction will be very important for structure based drug design. Lack of knowledge of 3D structures has hindered efforts to understand the binding specificities of ligands with protein. With increasing in modeling software and the growing number of known protein structures, homology modeling is rapidly becoming the method of choice for obtaining 3D coordinates of proteins. Homology modeling is a representation of the similarity of environmental residues at topologically corresponding positions in the reference proteins. In the absence of experimental data, model building on the basis of a known 3D structure of a homologous protein is at present the only reliable method to obtain the structural information. Knowledge of the 3D structures of proteins provides invaluable insights into the molecular basis of their functions. The recent advances in homology modeling, particularly in detecting and aligning sequences with template structures, distant homologues, modeling of loops and side chains as well as detecting errors in a model contributed to consistent prediction of protein structure, which was not possible even several years ago. This review focused on the features and a role of homology modeling in predicting protein structure and described current developments in this field with victorious applications at the different stages of the drug design and discovery.
Collapse
Affiliation(s)
- V K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad-382 481, India
| | | | | | | |
Collapse
|
43
|
Chow IT, James EA, Tan V, Moustakas AK, Papadopoulos GK, Kwok WW. DRB1*12:01 presents a unique subset of epitopes by preferring aromatics in pocket 9. Mol Immunol 2011; 50:26-34. [PMID: 22196942 DOI: 10.1016/j.molimm.2011.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/22/2011] [Accepted: 11/29/2011] [Indexed: 01/26/2023]
Abstract
This study characterized the unique peptide-binding characteristics of HLA-DRB1*12:01 (DR1201), an allele studied in the context of various autoimmune diseases, using a peptide competition assay and structural modeling. After defining Influenza A/Puerto Rico/8/34 Matrix Protein M1 (H1MP) 40-54 as a DR1201 restricted epitope, the critical anchor residues within this sequence were confirmed by measuring the relative binding of peptides with non-conservative substitutions in competition with biotin labeled H1MP(40-54) peptide. Based on this information, a set of peptides was designed with single amino acid substitutions at these anchor positions. The overall peptide binding preferences for the DR1201 allele were deduced by incubating these peptides in competition with the reference H1MP(40-54) to determine the relative binding affinities of each to recombinant DR1201 protein. As expected, pocket 1 preferred methionine and aliphatic residues, and tolerated phenylalanine. Pocket 4 was mostly composed of hydrophobic residues, thereby preferentially accommodating aliphatic residues, but could also weakly accommodate lysine due to its slightly acidic environment. Pocket 6 accepted a wide range of amino acids because of the diverse residues that comprise this pocket. Pocket 9 accepted aliphatic and negatively charged amino acids, but showed a remarkable preference for aromatic residues due to the conformation of the pocket, which lacks the typical salt bridge between β57Asp and α76Arg. These binding characteristics contrast with the closely related DR1104 allele, distinguishing DR1201 among the alleles of the HLA-DR5 group. These empirical results were used to develop an algorithm to predict peptide binding to DR1201. This algorithm was used to verify T cell epitopes within novel antigenic peptides identified by tetramer staining and within peptides from published reports that contain putative DR1201 epitopes.
Collapse
Affiliation(s)
- I-Ting Chow
- Benaroya Research Institute at Virginia Mason, 1201 9th Avenue, Seattle, WA 98101, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Sundaramurthy P, Sreenivasan R, Shameer K, Gakkhar S, Sowdhamini R. HORIBALFRE program: Higher Order Residue Interactions Based ALgorithm for Fold REcognition. Bioinformation 2011; 7:352-9. [PMID: 22355236 PMCID: PMC3280490 DOI: 10.6026/97320630007352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 11/24/2011] [Indexed: 11/23/2022] Open
Abstract
Understanding the functional and structural implication of a protein encoded in novel genes using function association or fold recognition approaches remains to be a challenging task in the current era of genomes, metagenomes and personal genomes. In an attempt to enhance potential-based fold-recognition methods in recognizing remote homology between proteins, we propose a new approach "Higher Order Residue Interaction Based ALgorithm for Fold REcognition (HORIBALFRE)". Higher order residue interactions refer to a class of interactions in protein structures mediated by C(α) or C(β) atoms within a pre-defined distance cut-off. Higher order residue interactions (pairwise, triplet and quadruplet interactions) play a vital role in attaining the stable conformation of a protein structure. In HORIBALFRE, we incorporated the potential contributions from two body (pairwise) interactions, three body (triplet interactions) and four-body (quadruple interaction) interactions, to implement a new fold recognition algorithm. Core of HORIBALFRE algorithm includes the potentials generated from a library of protein structure derived from manually curated CAMPASS database of structure based sequence alignment. We used Fischer's dataset, with 68 templates and 56 target sequences, derived from SCOP database and performed one-against-all sequence alignment using TCoffee. Various potentials were derived using custom scripts and these potentials were incorporated in the HORIBALFRE algorithm. In this manuscript, we report outline of a novel fold recognition algorithm and initial results. Our results show that inclusion of quadruplet class of higher order residue interaction improves fold recognition.
Collapse
Affiliation(s)
- Pandurangan Sundaramurthy
- National Center for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bellary Road, Bangalore - 560065, India
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee -247667, India
| | - Raashi Sreenivasan
- National Center for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bellary Road, Bangalore - 560065, India
- Centre for Biotechnology, Anna University, Chennai - 600025, India
- University of Wisconsin-Madison, Madison, WI 53706-1481, USA; 5Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55901 USA
| | - Khader Shameer
- National Center for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bellary Road, Bangalore - 560065, India
- Authors contributed equally to this work
| | - Sunita Gakkhar
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee -247667, India
| | - Ramanathan Sowdhamini
- National Center for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bellary Road, Bangalore - 560065, India
| |
Collapse
|
45
|
Shameer K, Shingate PN, Manjunath SCP, Karthika M, Pugalenthi G, Sowdhamini R. 3DSwap: curated knowledgebase of proteins involved in 3D domain swapping. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2011; 2011:bar042. [PMID: 21959866 PMCID: PMC3294423 DOI: 10.1093/database/bar042] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Three-dimensional domain swapping is a unique protein structural phenomenon where two or more protein chains in a protein oligomer share a common structural segment between individual chains. This phenomenon is observed in an array of protein structures in oligomeric conformation. Protein structures in swapped conformations perform diverse functional roles and are also associated with deposition diseases in humans. We have performed in-depth literature curation and structural bioinformatics analyses to develop an integrated knowledgebase of proteins involved in 3D domain swapping. The hallmark of 3D domain swapping is the presence of distinct structural segments such as the hinge and swapped regions. We have curated the literature to delineate the boundaries of these regions. In addition, we have defined several new concepts like ‘secondary major interface’ to represent the interface properties arising as a result of 3D domain swapping, and a new quantitative measure for the ‘extent of swapping’ in structures. The catalog of proteins reported in 3DSwap knowledgebase has been generated using an integrated structural bioinformatics workflow of database searches, literature curation, by structure visualization and sequence–structure–function analyses. The current version of the 3DSwap knowledgebase reports 293 protein structures, the analysis of such a compendium of protein structures will further the understanding molecular factors driving 3D domain swapping. Database URL:http://caps.ncbs.res.in/3dswap
Collapse
Affiliation(s)
- Khader Shameer
- National Centre for Biological Sciences, GKVK Campus, Bangalore, Karnataka, India
| | | | | | | | | | | |
Collapse
|
46
|
Shameer K, Madan LL, Veeranna S, Gopal B, Sowdhamini R. PeptideMine--a webserver for the design of peptides for protein-peptide binding studies derived from protein-protein interactomes. BMC Bioinformatics 2010; 11:473. [PMID: 20858292 PMCID: PMC2955050 DOI: 10.1186/1471-2105-11-473] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2009] [Accepted: 09/22/2010] [Indexed: 01/18/2023] Open
Abstract
Background Signal transduction events often involve transient, yet specific, interactions between structurally conserved protein domains and polypeptide sequences in target proteins. The identification and validation of these associating domains is crucial to understand signal transduction pathways that modulate different cellular or developmental processes. Bioinformatics strategies to extract and integrate information from diverse sources have been shown to facilitate the experimental design to understand complex biological events. These methods, primarily based on information from high-throughput experiments, have also led to the identification of new connections thus providing hypothetical models for cellular events. Such models, in turn, provide a framework for directing experimental efforts for validating the predicted molecular rationale for complex cellular processes. In this context, it is envisaged that the rational design of peptides for protein-peptide binding studies could substantially facilitate the experimental strategies to evaluate a predicted interaction. This rational design procedure involves the integration of protein-protein interaction data, gene ontology, physico-chemical calculations, domain-domain interaction data and information on functional sites or critical residues. Results Here we describe an integrated approach called "PeptideMine" for the identification of peptides based on specific functional patterns present in the sequence of an interacting protein. This approach based on sequence searches in the interacting sequence space has been developed into a webserver, which can be used for the identification and analysis of peptides, peptide homologues or functional patterns from the interacting sequence space of a protein. To further facilitate experimental validation, the PeptideMine webserver also provides a list of physico-chemical parameters corresponding to the peptide to determine the feasibility of using the peptide for in vitro biochemical or biophysical studies. Conclusions The strategy described here involves the integration of data and tools to identify potential interacting partners for a protein and design criteria for peptides based on desired biochemical properties. Alongside the search for interacting protein sequences using three different search programs, the server also provides the biochemical characteristics of candidate peptides to prune peptide sequences based on features that are most suited for a given experiment. The PeptideMine server is available at the URL: http://caps.ncbs.res.in/peptidemine
Collapse
Affiliation(s)
- Khader Shameer
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bellary Road, Bangalore, 560065, India
| | | | | | | | | |
Collapse
|
47
|
Meshram RJ, Gavhane A, Gaikar R, Bansode T, Maskar A, Gupta A, Sohni S, Patidar M, Pandey T, Jangle S. Sequence analysis and homology modeling of laccase from Pycnoporus cinnabarinus. Bioinformation 2010; 5:150-4. [PMID: 21364777 PMCID: PMC3040475 DOI: 10.6026/97320630005150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 08/26/2010] [Indexed: 11/30/2022] Open
Abstract
Industrial effluents of textile, paper, and leather industries contain various toxic dyes as one of the waste material. It imparts major impact on human
health as well as environment. The white rot fungus Pycnoporus cinnabarinus Laccase is generally used to degrade these toxic dyes. In order to decipher
the mechanism of process by which Laccase degrade dyes, it is essential to know its 3D structure. Homology modeling was performed in presented work,
by satisfying Spatial restrains using Modeller Program, which is considered as standard in this field, to generate 3D structure of Laccase in unison,
SWISSMODEL web server was also utilized to generate and verify the alternative models. We observed that models created using Modeller stands better
on structure evaluation tests. This study can further be used in molecular docking techniques, to understand the interaction of enzyme with its mediators
like 2, 2‐azinobis (3‐ethylbenzthiazoline‐6‐sulfonate) (ABTS) and Vanillin that are known to enhance the Laccase activity.
Collapse
Affiliation(s)
- Rohan J Meshram
- Center for Biotechnology, Pravara Institute of Medical Sciences, Loni, Taluka: Rahata, District: Ahmednagar, Maharashtra, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sundaramurthy P, Shameer K, Sreenivasan R, Gakkhar S, Sowdhamini R. HORI: a web server to compute Higher Order Residue Interactions in protein structures. BMC Bioinformatics 2010; 11 Suppl 1:S24. [PMID: 20122196 PMCID: PMC3009495 DOI: 10.1186/1471-2105-11-s1-s24] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Folding of a protein into its three dimensional structure is influenced by both local and global interactions within a protein. Higher order residue interactions, like pairwise, triplet and quadruplet ones, play a vital role in attaining the stable conformation of the protein structure. It is generally agreed that higher order interactions make significant contribution to the potential energy landscape of folded proteins and therefore it is important to identify them to estimate their contributions to overall stability of a protein structure. RESULTS We developed HORI [Higher order residue interactions in proteins], a web server for the calculation of global and local higher order interactions in protein structures. The basic algorithm of HORI is designed based on the classical concept of four-body nearest-neighbour propensities of amino-acid residues. It has been proved that higher order residue interactions up to the level of quadruple interactions plays a major role in the three-dimensional structure of proteins and is an important feature that can be used in protein structure analysis. CONCLUSION HORI server will be a useful resource for the structural bioinformatics community to perform analysis on protein structures based on higher order residue interactions. HORI server is a highly interactive web server designed in three modules that enables the user to analyse higher order residue interactions in protein structures. HORI server is available from the URL: http://caps.ncbs.res.in/hori.
Collapse
Affiliation(s)
- Pandurangan Sundaramurthy
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bellary Road, Bangalore, 560065, India.
| | | | | | | | | |
Collapse
|
49
|
Mijajlovic M, Biggs MJ, Djurdjevic DP. On potential energy models for EA-based ab initio protein structure prediction. EVOLUTIONARY COMPUTATION 2010; 18:255-275. [PMID: 20210597 DOI: 10.1162/evco.2010.18.2.18204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Ab initio protein structure prediction involves determination of the three-dimensional (3D) conformation of proteins on the basis of their amino acid sequence, a potential energy (PE) model that captures the physics of the interatomic interactions, and a method to search for and identify the global minimum in the PE (or free energy) surface such as an evolutionary algorithm (EA). Many PE models have been proposed over the past three decades and more. There is currently no understanding of how the behavior of an EA is affected by the PE model used. The study reported here shows that the EA behavior can be profoundly affected: the EA performance obtained when using the ECEPP PE model is significantly worse than that obtained when using the Amber, OPLS, and CVFF PE models, and the optimal EA control parameter values for the ECEPP model also differ significantly from those associated with the other models.
Collapse
Affiliation(s)
- Milan Mijajlovic
- Exobiology Branch, NASA Ames Research Center, Mail-Stop 239-4, Moffett Field, California 94035, USA
| | | | | |
Collapse
|
50
|
|