1
|
Katchkovsky S, Meiri R, Lacham‐Hartman S, Orenstein Y, Levaot N, Papo N. Mapping the sclerostin-LRP4 binding interface identifies critical interaction hotspots in loops 1 and 3 of sclerostin. FEBS Lett 2025; 599:316-329. [PMID: 39443289 PMCID: PMC11808424 DOI: 10.1002/1873-3468.15033] [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: 07/03/2024] [Revised: 08/19/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
The interaction of sclerostin (Scl) with the low-density lipoprotein receptor-related protein 4 (LRP4) leads to a marked reduction in bone formation by inhibiting the Wnt/β-catenin pathway. To characterize the Scl-LRP4 binding interface, we sorted a combinatorial library of Scl variants and isolated variants with reduced affinity to LRP4. We identified Scl single-mutation variants enriched during the sorting process and verified their reduction in affinity toward LRP4-a reduction that was not a result of changes in the variants' secondary structure or stability. We found that Scl positions K75 (loop 1) and V136 (loop 3) are critical hotspots for binding to LRP4. Our findings establish the foundation for targeting these hotspots for developing novel therapeutic strategies to promote bone formation.
Collapse
Affiliation(s)
- Svetlana Katchkovsky
- Department of Physiology and Cell Biology, Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Reut Meiri
- Department of Computer ScienceBar‐Ilan UniversityRamat GanIsrael
| | - Shiran Lacham‐Hartman
- Avram and Stella Goldstein‐Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the NegevBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Yaron Orenstein
- Department of Computer ScienceBar‐Ilan UniversityRamat GanIsrael
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat GanIsrael
| | - Noam Levaot
- Department of Physiology and Cell Biology, Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Niv Papo
- Avram and Stella Goldstein‐Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the NegevBen‐Gurion University of the NegevBeer‐ShevaIsrael
| |
Collapse
|
2
|
Xiao SR, Zhang YK, Liu KY, Huang YX, Liu R. PNBACE: an ensemble algorithm to predict the effects of mutations on protein-nucleic acid binding affinity. BMC Biol 2024; 22:203. [PMID: 39256728 PMCID: PMC11389284 DOI: 10.1186/s12915-024-02006-9] [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: 12/24/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Mutations occurring in nucleic acids or proteins may affect the binding affinities of protein-nucleic acid interactions. Although many efforts have been devoted to the impact of protein mutations, few computational studies have addressed the effect of nucleic acid mutations and explored whether the identical methodology could be applied to the prediction of binding affinity changes caused by these two mutation types. RESULTS Here, we developed a generalized algorithm named PNBACE for both DNA and protein mutations. We first demonstrated that DNA mutations could induce varying degrees of changes in binding affinity from multiple perspectives. We then designed a group of energy-based topological features based on different energy networks, which were combined with our previous partition-based energy features to construct individual prediction models through feature selections. Furthermore, we created an ensemble model by integrating the outputs of individual models using a differential evolution algorithm. In addition to predicting the impact of single-point mutations, PNBACE could predict the influence of multiple-point mutations and identify mutations significantly reducing binding affinities. Extensive comparisons indicated that PNBACE largely performed better than existing methods on both regression and classification tasks. CONCLUSIONS PNBACE is an effective method for estimating the binding affinity changes of protein-nucleic acid complexes induced by DNA or protein mutations, therefore improving our understanding of the interactions between proteins and DNA/RNA.
Collapse
Affiliation(s)
- Si-Rui Xiao
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yao-Kun Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Kai-Yu Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu-Xiang Huang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Rong Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
3
|
Islam S, Parves MR, Islam MJ, Ali MA, Efaz FM, Hossain MS, Ullah MO, Halim MA. Structural and functional effects of the L84S mutant in the SARS-COV-2 ORF8 dimer based on microsecond molecular dynamics study. J Biomol Struct Dyn 2024; 42:5770-5787. [PMID: 37403295 DOI: 10.1080/07391102.2023.2228919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
The L84S mutation has been observed frequently in the ORF8 protein of SARS-CoV-2, which is an accessory protein involved in various important functions such as virus propagation, pathogenesis, and evading the immune response. However, the specific effects of this mutation on the dimeric structure of ORF8 and its impacts on interactions with host components and immune responses are not well understood. In this study, we performed one microsecond molecular dynamics (MD) simulation and analyzed the dimeric behavior of the L84S and L84A mutants in comparison to the native protein. The MD simulations revealed that both mutations caused changes in the conformation of the ORF8 dimer, influenced protein folding mechanisms, and affected the overall structural stability. In particular, the 73YIDI76 motif has found to be significantly affected by the L84S mutation, leading to structural flexibility in the region connecting the C-terminal β4 and β5 strands. This flexibility might be responsible for virus immune modulation. The free energy landscape (FEL) and principle component analysis (PCA) have also supported our investigation. Overall, the L84S and L84A mutations affect the ORF8 dimeric interfaces by reducing the frequency of protein-protein interacting residues (Arg52, Lys53, Arg98, Ile104, Arg115, Val117, Asp119, Phe120, and Ile121) in the ORF8 dimer. Our findings provide detail insights for further research in designing structure-based therapeutics against the SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shafiqul Islam
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Rimon Parves
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Jahirul Islam
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Ackas Ali
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA, USA
| | - Faiyaz Md Efaz
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Shahadat Hossain
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - M Obayed Ullah
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Mohammad A Halim
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA, USA
| |
Collapse
|
4
|
Tao L, Zhou T, Wu Z, Hu F, Yang S, Kong X, Li C. ESPDHot: An Effective Machine Learning-Based Approach for Predicting Protein-DNA Interaction Hotspots. J Chem Inf Model 2024; 64:3548-3557. [PMID: 38587997 DOI: 10.1021/acs.jcim.3c02011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Protein-DNA interactions are pivotal to various cellular processes. Precise identification of the hotspot residues for protein-DNA interactions holds great significance for revealing the intricate mechanisms in protein-DNA recognition and for providing essential guidance for protein engineering. Aiming at protein-DNA interaction hotspots, this work introduces an effective prediction method, ESPDHot based on a stacked ensemble machine learning framework. Here, the interface residue whose mutation leads to a binding free energy change (ΔΔG) exceeding 2 kcal/mol is defined as a hotspot. To tackle the imbalanced data set issue, the adaptive synthetic sampling (ADASYN), an oversampling technique, is adopted to synthetically generate new minority samples, thereby rectifying data imbalance. As for molecular characteristics, besides traditional features, we introduce three new characteristic types including residue interface preference proposed by us, residue fluctuation dynamics characteristics, and coevolutionary features. Combining the Boruta method with our previously developed Random Grouping strategy, we obtained an optimal set of features. Finally, a stacking classifier is constructed to output prediction results, which integrates three classical predictors, Support Vector Machine (SVM), XGBoost, and Artificial Neural Network (ANN) as the first layer, and Logistic Regression (LR) algorithm as the second one. Notably, ESPDHot outperforms the current state-of-the-art predictors, achieving superior performance on the independent test data set, with F1, MCC, and AUC reaching 0.571, 0.516, and 0.870, respectively.
Collapse
Affiliation(s)
- Lianci Tao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Tong Zhou
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Zhixiang Wu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Fangrui Hu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Shuang Yang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xiaotian Kong
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Chunhua Li
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
5
|
Crean RM, Pudney CR, Cole DK, van der Kamp MW. Reliable In Silico Ranking of Engineered Therapeutic TCR Binding Affinities with MMPB/GBSA. J Chem Inf Model 2022; 62:577-590. [PMID: 35049312 PMCID: PMC9097153 DOI: 10.1021/acs.jcim.1c00765] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Accurate
and efficient in silico ranking of protein–protein
binding affinities is useful for protein design with applications
in biological therapeutics. One popular approach to rank binding affinities
is to apply the molecular mechanics Poisson–Boltzmann/generalized
Born surface area (MMPB/GBSA) method to molecular dynamics (MD) trajectories.
Here, we identify protocols that enable the reliable evaluation of
T-cell receptor (TCR) variants binding to their target, peptide-human
leukocyte antigens (pHLAs). We suggest different protocols for variant
sets with a few (≤4) or many mutations, with entropy corrections
important for the latter. We demonstrate how potential outliers could
be identified in advance and that just 5–10 replicas of short
(4 ns) MD simulations may be sufficient for the reproducible and accurate
ranking of TCR variants. The protocols developed here can be applied
toward in silico screening during the optimization
of therapeutic TCRs, potentially reducing both the cost and time taken
for biologic development.
Collapse
Affiliation(s)
| | | | - David K. Cole
- Immunocore Ltd., Milton Park, Abingdon OX14 4RY, U.K
- Division of Infection & Immunity, Cardiff University, Cardiff CF14 4XN, U.K
| | - Marc W. van der Kamp
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, U.K
| |
Collapse
|
6
|
Investigation of glutathione as a natural antioxidant and multitarget inhibitor for Alzheimer’s disease: Insights from molecular simulations. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
Hydrophobic Residues Confer the Helicity and Membrane Permeability of Ocellatin-1 Antimicrobial Peptide Scaffold Towards Therapeutics. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10265-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
8
|
Corbella M, Liao Q, Moreira C, Parracino A, Kasson PM, Kamerlin SCL. The N-terminal Helix-Turn-Helix Motif of Transcription Factors MarA and Rob Drives DNA Recognition. J Phys Chem B 2021; 125:6791-6806. [PMID: 34137249 PMCID: PMC8279559 DOI: 10.1021/acs.jpcb.1c00771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
DNA-binding proteins
play an important role in gene regulation
and cellular function. The transcription factors MarA and Rob are
two homologous members of the AraC/XylS family that regulate multidrug
resistance. They share a common DNA-binding domain, and Rob possesses
an additional C-terminal domain that permits binding of low-molecular
weight effectors. Both proteins possess two helix-turn-helix (HTH)
motifs capable of binding DNA; however, while MarA interacts with
its promoter through both HTH-motifs, prior studies indicate that
Rob binding to DNA via a single HTH-motif is sufficient for tight
binding. In the present work, we perform microsecond time scale all-atom
simulations of the binding of both transcription factors to different
DNA sequences to understand the determinants of DNA recognition and
binding. Our simulations characterize sequence-dependent changes in
dynamical behavior upon DNA binding, showcasing the role of Arg40
of the N-terminal HTH-motif in allowing for specific tight binding.
Finally, our simulations demonstrate that an acidic C-terminal loop
of Rob can control the DNA binding mode, facilitating interconversion
between the distinct DNA binding modes observed in MarA and Rob. In
doing so, we provide detailed molecular insight into DNA binding and
recognition by these proteins, which in turn is an important step
toward the efficient design of antivirulence agents that target these
proteins.
Collapse
Affiliation(s)
- Marina Corbella
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Uppsala, S-751 23, Sweden
| | - Qinghua Liao
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Uppsala, S-751 23, Sweden
| | - Cátia Moreira
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Uppsala, S-751 23, Sweden
| | - Antonietta Parracino
- Science for Life Laboratory, Department of Chemistry-BMC, Uppsala University, Uppsala, S-751 23, Sweden
| | - Peter M Kasson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, S-65124, Sweden.,Departments of Molecular Physiology and Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | | |
Collapse
|
9
|
Vračko M, Basak SC, Sen D, Nandy A. Clustering of Zika Viruses Originating from Different Geographical Regions using Computational Sequence Descriptors. Curr Comput Aided Drug Des 2021; 17:314-322. [PMID: 31878862 DOI: 10.2174/1573409916666191226110936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/18/2019] [Accepted: 12/09/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND In this report, we consider a data set, which consists of 310 Zika virus genome sequences taken from different continents, Africa, Asia and South America. The sequences, which were compiled from GenBank, were derived from the host cells of different mammalian species (Simiiformes, Aedes opok, Aedes africanus, Aedes luteocephalus, Aedes dalzieli, Aedes aegypti, and Homo sapiens). METHODS For chemometrical treatment, the sequences have been represented by sequence descriptors derived from their graphs or neighborhood matrices. The set was analyzed with three chemometrical methods: Mahalanobis distances, principal component analysis (PCA) and self organizing maps (SOM). A good separation of samples with respect to the region of origin was observed using these three methods. RESULTS Study of 310 Zika virus genome sequences from different continents. To characterize and compare Zika virus sequences from around the world using alignment-free sequence comparison and chemometrical methods. CONCLUSION Mahalanobis distance analysis, self organizing maps, principal components were used to carry out the chemometrical analyses of the Zika sequence data. Genome sequences are clustered with respect to the region of origin (continent, country). Africa samples are well separated from Asian and South American ones.
Collapse
Affiliation(s)
- Marjan Vračko
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Subhash C Basak
- Department of Chemistry and Biochemistry, University of Minnesota, Duluth, United States
| | - Dwaipayan Sen
- Centre for Interdisciplinary Research and Education, Kolkata, India
| | - Ashesh Nandy
- Centre for Interdisciplinary Research and Education, Kolkata, India
| |
Collapse
|
10
|
Alizadeh AA, Dastmalchi S. Designing Novel Teduglutide Analogues with Improved Binding Affinity: An In Silico Peptide Engineering Approach. Curr Comput Aided Drug Des 2021; 17:225-234. [PMID: 32065094 DOI: 10.2174/1573409916666200217091456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/05/2019] [Accepted: 01/17/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Short bowel syndrome (SBS) is a disabling condition that occurs following the loss of substantial portions of the intestine, leading to inadequate absorption of nutrients and fluids. Teduglutide is the only drug that has been FDA-approved for long-term treatment of SBS. This medicine exerts its biological effects through binding to the GLP-2 receptor. METHODS The current study aimed to use computational mutagenesis approaches to design novel potent analogues of teduglutide. To this end, the constructed teduglutide-GLP2R 3D model was subjected to the alanine scanning mutagenesis where ARG20, PHE22, ILE23, LEU26, ILE27 and LYS30 were identified as the key amino acids involved in ligand-receptor interaction. In order to design potent teduglutide analogues, using MAESTROweb machine learning method, the residues of teduglutide were virtually mutated into all naturally occurring amino acids and the affinity improving mutations were selected for further analysis using PDBePISA methodology which interactively investigates the interactions established at the interfaces of macromolecules. RESULTS The calculations resulted in D15I, D15L, D15M and N24M mutations, which can improve the binding ability of the ligand to the receptor. The final evaluation of identified mutations was performed by molecular dynamics simulations, indicating that D15I and D15M are the most reliable mutations to increase teduglutide affinity towards its receptor. CONCLUSION The findings in the current study may facilitate designing more potent teduglutide analogues leading to the development of novel treatments in short bowel syndrome.
Collapse
Affiliation(s)
- Ali A Alizadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Jiang Y, Liu HF, Liu R. Systematic comparison and prediction of the effects of missense mutations on protein-DNA and protein-RNA interactions. PLoS Comput Biol 2021; 17:e1008951. [PMID: 33872313 PMCID: PMC8084330 DOI: 10.1371/journal.pcbi.1008951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/29/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
The binding affinities of protein-nucleic acid interactions could be altered due to missense mutations occurring in DNA- or RNA-binding proteins, therefore resulting in various diseases. Unfortunately, a systematic comparison and prediction of the effects of mutations on protein-DNA and protein-RNA interactions (these two mutation classes are termed MPDs and MPRs, respectively) is still lacking. Here, we demonstrated that these two classes of mutations could generate similar or different tendencies for binding free energy changes in terms of the properties of mutated residues. We then developed regression algorithms separately for MPDs and MPRs by introducing novel geometric partition-based energy features and interface-based structural features. Through feature selection and ensemble learning, similar computational frameworks that integrated energy- and nonenergy-based models were established to estimate the binding affinity changes resulting from MPDs and MPRs, but the selected features for the final models were different and therefore reflected the specificity of these two mutation classes. Furthermore, the proposed methodology was extended to the identification of mutations that significantly decreased the binding affinities. Extensive validations indicated that our algorithm generally performed better than the state-of-the-art methods on both the regression and classification tasks. The webserver and software are freely available at http://liulab.hzau.edu.cn/PEMPNI and https://github.com/hzau-liulab/PEMPNI. Protein-nucleic acid interactions play important roles in various cellular processes. Missense mutations occurring in DNA- or RNA-binding proteins (termed MPDs and MPRs, respectively) could change the binding affinities of these interactions. Previous studies have compared protein-DNA and protein-RNA interactions from multifaceted viewpoints, but less attention has been given to the similarities and specific differences between the effects of MPDs and MPRs and between the methodologies for predicting the affinity changes induced by the two mutation classes. Therefore, we systematically compared their impacts and demonstrated that MPDs and MPRs could have specific preferences for binding affinity changes. These observations motivated us to construct regression models separately for MPDs and MPRs by introducing novel energy and nonenergy descriptors. Although similar frameworks were developed to estimate these two categories of mutation effects, different descriptors were selected in the regression models and further revealed the specificity of mutation classes. The interplay between the energy and nonenergy modules effectively improved prediction performance. Our algorithm can also be adopted to disentangle mutations significantly decreasing binding affinities from other mutations.
Collapse
Affiliation(s)
- Yao Jiang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
| | - Hui-Fang Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
| | - Rong Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
| |
Collapse
|
12
|
Hassanzadeh K, Perez Pena H, Dragotto J, Buccarello L, Iorio F, Pieraccini S, Sancini G, Feligioni M. Considerations around the SARS-CoV-2 Spike Protein with Particular Attention to COVID-19 Brain Infection and Neurological Symptoms. ACS Chem Neurosci 2020; 11:2361-2369. [PMID: 32627524 PMCID: PMC7374936 DOI: 10.1021/acschemneuro.0c00373] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
Spike protein (S protein) is the virus "key" to infect cells and is able to strongly bind to the human angiotensin-converting enzyme2 (ACE2), as has been reported. In fact, Spike structure and function is known to be highly important for cell infection as well as for entering the brain. Growing evidence indicates that different types of coronaviruses not only affect the respiratory system, but they might also invade the central nervous system (CNS). However, very little evidence has been so far reported on the presence of COVID-19 in the brain, and the potential exploitation, by this virus, of the lung to brain axis to reach neurons has not been completely understood. In this Article, we assessed the SARS-CoV and SARS-CoV-2 Spike protein sequence, structure, and electrostatic potential using computational approaches. Our results showed that the S proteins of SARS-CoV-2 and SARS-CoV are highly similar, sharing a sequence identity of 77%. In addition, we found that the SARS-CoV-2 S protein is slightly more positively charged than that of SARS-CoV since it contains four more positively charged residues and five less negatively charged residues which may lead to an increased affinity to bind to negatively charged regions of other molecules through nonspecific and specific interactions. Analysis the S protein binding to the host ACE2 receptor showed a 30% higher binding energy for SARS-CoV-2 than for the SARS-CoV S protein. These results might be useful for understanding the mechanism of cell entry, blood-brain barrier crossing, and clinical features related to the CNS infection by SARS-CoV-2.
Collapse
Affiliation(s)
- Kambiz Hassanzadeh
- Laboratory of Neuronal Cell Signaling,
EBRI Rita Levi-Montalcini Foundation, Rome 00161,
Italy
- Cellular and Molecular Research Center, Research
Institute for Health Development, Kurdistan University of Medical
Sciences, Sanandaj 66177-13446, Iran
| | - Helena Perez Pena
- Department of Chemistry and National Inter-University
Consortium for Materials Science and Technology-INSTM-UdR Milano, University
of Milan, Milan 20133, Italy
| | - Jessica Dragotto
- Laboratory of Neuronal Cell Signaling,
EBRI Rita Levi-Montalcini Foundation, Rome 00161,
Italy
| | - Lucia Buccarello
- Laboratory of Neuronal Cell Signaling,
EBRI Rita Levi-Montalcini Foundation, Rome 00161,
Italy
| | - Federico Iorio
- Laboratory of Neuronal Cell Signaling,
EBRI Rita Levi-Montalcini Foundation, Rome 00161,
Italy
| | - Stefano Pieraccini
- Department of Chemistry and National Inter-University
Consortium for Materials Science and Technology-INSTM-UdR Milano, University
of Milan, Milan 20133, Italy
- Institute of Science and Chemical
Technology “Giulio Natta”, Milan 20133,
Italy
| | - Giulio Sancini
- Human Physiology Lab, School of Medicine and Surgery,
University of Milano-Bicocca, Via Cadore 48, 20900 Monza,
Italy
| | - Marco Feligioni
- Laboratory of Neuronal Cell Signaling,
EBRI Rita Levi-Montalcini Foundation, Rome 00161,
Italy
- Department of Neurorehabilitation Sciences, Casa
di Cura del Policlinico, Milan 20144, Italy
| |
Collapse
|
13
|
Zhu X, Liu L, He J, Fang T, Xiong Y, Mitchell JC. iPNHOT: a knowledge-based approach for identifying protein-nucleic acid interaction hot spots. BMC Bioinformatics 2020; 21:289. [PMID: 32631222 PMCID: PMC7336410 DOI: 10.1186/s12859-020-03636-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/25/2020] [Indexed: 12/20/2022] Open
Abstract
Background The interaction between proteins and nucleic acids plays pivotal roles in various biological processes such as transcription, translation, and gene regulation. Hot spots are a small set of residues that contribute most to the binding affinity of a protein-nucleic acid interaction. Compared to the extensive studies of the hot spots on protein-protein interfaces, the hot spot residues within protein-nucleic acids interfaces remain less well-studied, in part because mutagenesis data for protein-nucleic acids interaction are not as abundant as that for protein-protein interactions. Results In this study, we built a new computational model, iPNHOT, to effectively predict hot spot residues on protein-nucleic acids interfaces. One training data set and an independent test set were collected from dbAMEPNI and some recent literature, respectively. To build our model, we generated 97 different sequential and structural features and used a two-step strategy to select the relevant features. The final model was built based only on 7 features using a support vector machine (SVM). The features include two unique features such as ∆SASsa1/2 and esp3, which are newly proposed in this study. Based on the cross validation results, our model gave F1 score and AUROC as 0.725 and 0.807 on the subset collected from ProNIT, respectively, compared to 0.407 and 0.670 of mCSM-NA, a state-of-the art model to predict the thermodynamic effects of protein-nucleic acid interaction. The iPNHOT model was further tested on the independent test set, which showed that our model outperformed other methods. Conclusion In this study, by collecting data from a recently published database dbAMEPNI, we proposed a new model, iPNHOT, to predict hotspots on both protein-DNA and protein-RNA interfaces. The results show that our model outperforms the existing state-of-art models. Our model is available for users through a webserver: http://zhulab.ahu.edu.cn/iPNHOT/.
Collapse
Affiliation(s)
- Xiaolei Zhu
- School of Sciences, Anhui Agricultural University, Hefei, Anhui, China. .,School of Life Sciences, Anhui University, Hefei, Anhui, China.
| | - Ling Liu
- School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Jingjing He
- School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Ting Fang
- School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Yi Xiong
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Julie C Mitchell
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
| |
Collapse
|
14
|
Poli G, Granchi C, Rizzolio F, Tuccinardi T. Application of MM-PBSA Methods in Virtual Screening. Molecules 2020; 25:molecules25081971. [PMID: 32340232 PMCID: PMC7221544 DOI: 10.3390/molecules25081971] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
Computer-aided drug design techniques are today largely applied in medicinal chemistry. In particular, receptor-based virtual screening (VS) studies, in which molecular docking represents the gold standard in silico approach, constitute a powerful strategy for identifying novel hit compounds active against the desired target receptor. Nevertheless, the need for improving the ability of docking in discriminating true active ligands from inactive compounds, thus boosting VS hit rates, is still pressing. In this context, the use of binding free energy evaluation approaches can represent a profitable tool for rescoring ligand-protein complexes predicted by docking based on more reliable estimations of ligand-protein binding affinities than those obtained with simple scoring functions. In the present review, we focused our attention on the Molecular Mechanics-Poisson Boltzman Surface Area (MM-PBSA) method for the calculation of binding free energies and its application in VS studies. We provided examples of successful applications of this method in VS campaigns and evaluation studies in which the reliability of this approach has been assessed, thus providing useful guidelines for employing this approach in VS.
Collapse
Affiliation(s)
- Giulio Poli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.P.); (C.G.)
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.P.); (C.G.)
| | - Flavio Rizzolio
- Department of Molecular science and Nanosystems, University Ca’ Foscari of Venice, 30170 Venice, Italy;
- Pathology unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.P.); (C.G.)
- Correspondence: ; Tel.: +39-0502219595
| |
Collapse
|
15
|
Tian S, Ji C, Zhang JZH. Molecular basis of SMAC-XIAP binding and the effect of electrostatic polarization. J Biomol Struct Dyn 2020; 39:743-752. [PMID: 31914860 DOI: 10.1080/07391102.2020.1713892] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
X-chromosome-linked inhibitor of apoptosis (XIAP) inhibits cell apoptosis. Overexpression of XIAP is widely found in human cancers. Second mitochondria-derived activator of caspase (SMAC) protein inhibits XIAP through binding with Baculovirus Inhibitor of apoptosis protein Repeat (BIR) 3 or BIR2 domain of XIAP. In this study, molecular dynamics (MD) simulations and the alanine scanning calculations by MM-GBSA_IE method were used to investigate the protein-peptide interaction between BIR3 and BIR2 domains of XIAP and SMAC peptide. Energetic contribution of each binding residue is calculated and hotspots on both XIAP and SMAC were identified using computational alanine scanning with interaction entropy method. We found that electrostatic polarization is important in stabilizing the protein-protein complex structure in MD simulation. By using polarized protein-specific charges, much better agreement with experimental result is obtained for calculated binding free energies compared to those using standard (nonpolarizable) AMBER force field. In particular, excellent correlation between calculated binding free energies in alanine scanning with mutational experimental data was obtained for BIR3/SMAC binding.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shuaizhen Tian
- State Key Laboratory of Precision Spectroscopy and Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China
| | - Changge Ji
- State Key Laboratory of Precision Spectroscopy and Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, China
| | - John Z H Zhang
- State Key Laboratory of Precision Spectroscopy and Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, China.,Department of Chemistry, New York University, New York, NY, USA.,Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi, China
| |
Collapse
|
16
|
Ahmed MH, Catalano C, Portillo SC, Safo MK, Neel Scarsdale J, Kellogg GE. 3D interaction homology: The hydropathic interaction environments of even alanine are diverse and provide novel structural insight. J Struct Biol 2019; 207:183-198. [DOI: 10.1016/j.jsb.2019.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/12/2019] [Accepted: 05/17/2019] [Indexed: 01/23/2023]
|
17
|
Junaid M, Li CD, Shah M, Khan A, Guo H, Wei DQ. Extraction of molecular features for the drug discovery targeting protein-protein interaction of Helicobacter pylori CagA and tumor suppressor protein ASSP2. Proteins 2019; 87:837-849. [PMID: 31134671 DOI: 10.1002/prot.25748] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/04/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022]
Abstract
Half of the world population is infected by the Gram-negative bacterium Helicobacter pylori (H. pylori). It colonizes in the stomach and is associated with severe gastric pathologies including gastric cancer and peptic ulceration. The most virulent factor of H. pylori is the cytotoxin-associated gene A (CagA) that is injected into the host cell. CagA interacts with several host proteins and alters their function, thereby causing several diseases. The most well-known target of CagA is the tumor suppressor protein ASPP2. The subdomain I at the N-terminus of CagA interacts with the proline-rich motif of ASPP2. Here, in this study, we carried out alanine scanning mutagenesis and an extensive molecular dynamics simulation summing up to 3.8 μs to find out hot spot residues and discovered some new protein-protein interaction (PPI)-modulating molecules. Our findings are in line with previous biochemical studies and further suggested new residues that are crucial for binding. The alanine scanning showed that mutation of Y207 and T211 residues to alanine decreased the binding affinity. Likewise, dynamics simulation and molecular mechanics with generalized Born surface area (MMGBSA) analysis also showed the importance of these two residues at the interface. A four-feature pharmacophore model was developed based on these two residues, and top 10 molecules were filtered from ZINC, NCI, and ChEMBL databases. The good binding affinity of the CHEMBL17319 and CHEMBL1183979 molecules shows the reliability of our adopted protocol for binding hot spot residues. We believe that our study provides a new insight for using CagA as the therapeutic target for gastric cancer treatment and provides a platform for a future experimental study.
Collapse
Affiliation(s)
- Muhammad Junaid
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng-Dong Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Abbas Khan
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyue Guo
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Huang D, Wen W, Liu X, Li Y, Zhang JZH. Computational analysis of hot spots and binding mechanism in the PD-1/PD-L1 interaction. RSC Adv 2019; 9:14944-14956. [PMID: 35516311 PMCID: PMC9064197 DOI: 10.1039/c9ra01369e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/05/2019] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) is an important immunological checkpoint and plays a vital role in maintaining the peripheral tolerance of the human body by interacting with its ligand PD-L1. The overexpression of PD-L1 in tumor cells induces local immune suppression and helps the tumor cells to evade the endogenous anti-tumor immunity. Developing monoclonal antibodies against the PD-1/PD-L1 protein–protein interaction to block the PD-1/PD-L1 signaling pathway has demonstrated superior anti-tumor efficacy in a variety of solid tumors and has made a profound impact on the field of cancer immunotherapy in recent years. Although the X-ray crystal structure of the PD-1/PD-L1 complex has been solved, the detailed binding mechanism of the PD-1/PD-L1 interaction is not fully understood from a theoretical point of view. In this study, we performed computational alanine scanning on the PD-1/PD-L1 complex to quantitatively identify the hot spots in the PD-1/PD-L1 interaction and characterize its binding mechanisms at the atomic level. To the best of our knowledge, this is the first time that theoretical calculations have been used to systematically and quantitatively predict the hot spots in the PD-1/PD-L1 interaction. We hope that the predicted hot spots and the energy profile of the PD-1/PD-L1 interaction presented in this work can provide guidance for the design of peptide and small molecule drugs targeting PD-1 or PD-L1. The hot spots quantitatively predicted by the recently developed MM/GBSA/IE method reveal a hydrophobic core in the PD-1/PD-L1 interaction.![]()
Collapse
Affiliation(s)
- Dading Huang
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - Wei Wen
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - Xiao Liu
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - Yang Li
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| | - John Z. H. Zhang
- State Key Laboratory for Precision Spectroscopy
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
| |
Collapse
|
19
|
Huang D, Qi Y, Song J, Zhang JZH. Calculation of hot spots for protein–protein interaction in p53/PMI‐MDM2/MDMX complexes. J Comput Chem 2018; 40:1045-1056. [DOI: 10.1002/jcc.25592] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/04/2018] [Accepted: 08/23/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Dading Huang
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 China
| | - Yifei Qi
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 China
- NYU‐ECNU Center for Computational Chemistry at NYU Shanghai Shanghai 200062 China
| | - Jianing Song
- NYU‐ECNU Center for Computational Chemistry at NYU Shanghai Shanghai 200062 China
| | - John Z. H. Zhang
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 China
- NYU‐ECNU Center for Computational Chemistry at NYU Shanghai Shanghai 200062 China
- Department of ChemistryNew York University New York New York, 10003
- Collaborative Innovation Center of Extreme OpticsShanxi University Taiyuan Shanxi, 030006 China
| |
Collapse
|
20
|
Liu L, Xiong Y, Gao H, Wei DQ, Mitchell JC, Zhu X. dbAMEPNI: a database of alanine mutagenic effects for protein-nucleic acid interactions. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2018; 2018:4959188. [PMID: 29688380 PMCID: PMC5887268 DOI: 10.1093/database/bay034] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/15/2018] [Indexed: 01/08/2023]
Abstract
Protein–nucleic acid interactions play essential roles in various biological activities such as gene regulation, transcription, DNA repair and DNA packaging. Understanding the effects of amino acid substitutions on protein–nucleic acid binding affinities can help elucidate the molecular mechanism of protein–nucleic acid recognition. Until now, no comprehensive and updated database of quantitative binding data on alanine mutagenic effects for protein–nucleic acid interactions is publicly accessible. Thus, we developed a new database of Alanine Mutagenic Effects for Protein-Nucleic Acid Interactions (dbAMEPNI). dbAMEPNI is a manually curated, literature-derived database, comprising over 577 alanine mutagenic data with experimentally determined binding affinities for protein–nucleic acid complexes. It contains several important parameters, such as dissociation constant (Kd), Gibbs free energy change (ΔΔG), experimental conditions and structural parameters of mutant residues. In addition, the database provides an extended dataset of 282 single alanine mutations with only qualitative data (or descriptive effects) of thermodynamic information. Database URL: http://zhulab.ahu.edu.cn/dbAMEPNI
Collapse
Affiliation(s)
- Ling Liu
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongyun Gao
- Information and Engineering College, Dalian University, Dalian 116622, Liaoning, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Julie C Mitchell
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.,Department of Mathematics, University of Wisconsin-Madison, Madison, WI 53706, USA.,Oak Ridge National Laboratory, Biosciences Division, Oak Ridge, TN 37830, USA
| | - Xiaolei Zhu
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
21
|
Moreira IS, Koukos PI, Melo R, Almeida JG, Preto AJ, Schaarschmidt J, Trellet M, Gümüş ZH, Costa J, Bonvin AMJJ. SpotOn: High Accuracy Identification of Protein-Protein Interface Hot-Spots. Sci Rep 2017; 7:8007. [PMID: 28808256 PMCID: PMC5556074 DOI: 10.1038/s41598-017-08321-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022] Open
Abstract
We present SpotOn, a web server to identify and classify interfacial residues as Hot-Spots (HS) and Null-Spots (NS). SpotON implements a robust algorithm with a demonstrated accuracy of 0.95 and sensitivity of 0.98 on an independent test set. The predictor was developed using an ensemble machine learning approach with up-sampling of the minor class. It was trained on 53 complexes using various features, based on both protein 3D structure and sequence. The SpotOn web interface is freely available at: http://milou.science.uu.nl/services/SPOTON/.
Collapse
Affiliation(s)
- Irina S Moreira
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1°andar, Universidade de Coimbra, 3004-517, Coimbra, Portugal. .,Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht, 3584CH, The Netherlands.
| | - Panagiotis I Koukos
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht, 3584CH, The Netherlands
| | - Rita Melo
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1°andar, Universidade de Coimbra, 3004-517, Coimbra, Portugal.,Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (ao km 139,7), 2695-066, Bobadela LRS, Portugal
| | - Jose G Almeida
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1°andar, Universidade de Coimbra, 3004-517, Coimbra, Portugal
| | - Antonio J Preto
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1°andar, Universidade de Coimbra, 3004-517, Coimbra, Portugal
| | - Joerg Schaarschmidt
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht, 3584CH, The Netherlands
| | - Mikael Trellet
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht, 3584CH, The Netherlands
| | - Zeynep H Gümüş
- Department of Genetics and Genomics and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joaquim Costa
- CMUP/FCUP, Centro de Matemática da Universidade do Porto, Faculdade de Ciências, Rua do Campo Alegre, 4169-007, Porto, Portugal
| | - Alexandre M J J Bonvin
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht, 3584CH, The Netherlands.
| |
Collapse
|
22
|
Moktan H, Zhou DH. Wing 1 of protein HOP2 is as important as helix 3 in DNA binding by MD simulation. J Biomol Struct Dyn 2017; 36:1853-1866. [PMID: 28531371 DOI: 10.1080/07391102.2017.1333458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The repair of programmed DNA double-strand breaks through recombination is required for proper association and disjunction of the meiotic homologous chromosomes. Meiosis-specific protein HOP2 plays essential roles in recombination by promoting recombinase activities. The N-terminal domain of HOP2 interacts with DNA through helix 3 (H3) and wing 1 (W1). Mutations in wing 1 (Y65A/K67A/Q68A) slightly weakened the binding but mutations in helices 2 and 3 (Q30A/K44A/K49A) nearly abolished the binding. To better understand such differential effects at atomic level, molecular dynamics simulations were employed. Despite losing some hydrogen bonds, the W1-mutant DNA complex was rescued by stronger hydrophobic interactions. For the wild type and W1-mutant, the protein was found to slide along the DNA grooves as the DNA rolls along its double-helix axis. This motion could be functionally important to facilitate the precise positioning of the single-stranded DNA with the homologous double-stranded DNA. The sliding motion was reduced in the W1-mutant. The H-mutant nearly lost all intermolecular interactions. Moreover, an additional mutation in wing 1 (Y65A/K67A/Q68A/K69A) also caused complete complex dissociation. Therefore, both wing 1 and helix 3 make important contribution to the DNA binding, which could be important to the strand invasion function of HOP2 homodimer and HOP2-MND1 heterodimer. Similar to cocking a medieval crossbow with the archer's foot placed in the stirrup, wing 1 may push the minor groove to cause distortion while helix 3 grabs the major groove.
Collapse
Affiliation(s)
- Hem Moktan
- a Department of Physics , Oklahoma State University , Stillwater , OK , USA
| | - Donghua H Zhou
- a Department of Physics , Oklahoma State University , Stillwater , OK , USA
| |
Collapse
|
23
|
Zarei O, Hamzeh-Mivehroud M, Benvenuti S, Ustun-Alkan F, Dastmalchi S. Characterizing the Hot Spots Involved in RON-MSPβ Complex Formation Using In Silico Alanine Scanning Mutagenesis and Molecular Dynamics Simulation. Adv Pharm Bull 2017; 7:141-150. [PMID: 28507948 PMCID: PMC5426727 DOI: 10.15171/apb.2017.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 12/30/2022] Open
Abstract
Purpose: Implication of protein-protein interactions (PPIs) in development of many diseases such as cancer makes them attractive for therapeutic intervention and rational drug design. RON (Recepteur d'Origine Nantais) tyrosine kinase receptor has gained considerable attention as promising target in cancer therapy. The activation of RON via its ligand, macrophage stimulation protein (MSP) is the most common mechanism of activation for this receptor. The aim of the current study was to perform in silico alanine scanning mutagenesis and to calculate binding energy for prediction of hot spots in protein-protein interface between RON and MSPβ chain (MSPβ). Methods: In this work the residues at the interface of RON-MSPβ complex were mutated to alanine and then molecular dynamics simulation was used to calculate binding free energy. Results: The results revealed that Gln193, Arg220, Glu287, Pro288, Glu289, and His424 residues from RON and Arg521, His528, Ser565, Glu658, and Arg683 from MSPβ may play important roles in protein-protein interaction between RON and MSP. Conclusion: Identification of these RON hot spots is important in designing anti-RON drugs when the aim is to disrupt RON-MSP interaction. In the same way, the acquired information regarding the critical amino acids of MSPβ can be used in the process of rational drug design for developing MSP antagonizing agents, the development of novel MSP mimicking peptides where inhibition of RON activation is required, and the design of experimental site directed mutagenesis studies.
Collapse
Affiliation(s)
- Omid Zarei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Benvenuti
- Molecular Therapeutics and Exploratory Research Laboratory, Candiolo Cancer Institute-FPO-IRCCS, Candiolo, Turin, Italy
| | - Fulya Ustun-Alkan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, Turkey
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Bhandare VV, Ramaswamy A. The proteinopathy of D169G and K263E mutants at the RNA Recognition Motif (RRM) domain of tar DNA-binding protein (tdp43) causing neurological disorders: A computational study. J Biomol Struct Dyn 2017; 36:1075-1093. [DOI: 10.1080/07391102.2017.1310670] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
| | - Amutha Ramaswamy
- Centre for Bioinformatics, Pondicherry University, Pondicherry 605014, India
| |
Collapse
|
25
|
Lakhani B, Thayer KM, Hingorani MM, Beveridge DL. Evolutionary Covariance Combined with Molecular Dynamics Predicts a Framework for Allostery in the MutS DNA Mismatch Repair Protein. J Phys Chem B 2017; 121:2049-2061. [PMID: 28135092 PMCID: PMC5346969 DOI: 10.1021/acs.jpcb.6b11976] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
![]()
Mismatch
repair (MMR) is an essential, evolutionarily conserved
pathway that maintains genome stability by correcting base-pairing
errors in DNA. Here we examine the sequence and structure of MutS
MMR protein to decipher the amino acid framework underlying its two
key activities—recognizing mismatches in DNA and using ATP
to initiate repair. Statistical coupling analysis (SCA) identified
a network (sector) of coevolved amino acids in the MutS protein family.
The potential functional significance of this SCA sector was assessed
by performing molecular dynamics (MD) simulations for alanine mutants
of the top 5% of 160 residues in the distribution, and control nonsector
residues. The effects on three independent metrics were monitored:
(i) MutS domain conformational dynamics, (ii) hydrogen bonding between
MutS and DNA/ATP, and (iii) relative ATP binding free energy. Each
measure revealed that sector residues contribute more substantively
to MutS structure–function than nonsector residues. Notably,
sector mutations disrupted MutS contacts with DNA and/or ATP from
a distance via contiguous pathways and correlated motions, supporting
the idea that SCA can identify amino acid networks underlying allosteric
communication. The combined SCA/MD approach yielded novel, experimentally
testable hypotheses for unknown roles of many residues distributed
across MutS, including some implicated in Lynch cancer syndrome.
Collapse
Affiliation(s)
- Bharat Lakhani
- Molecular Biology and Biochemistry Department, ‡Molecular Biophysics Program, §Chemistry Department, and ∥Computer Science Department, Wesleyan University , Middletown, Connecticut 06459, United States
| | - Kelly M Thayer
- Molecular Biology and Biochemistry Department, ‡Molecular Biophysics Program, §Chemistry Department, and ∥Computer Science Department, Wesleyan University , Middletown, Connecticut 06459, United States
| | - Manju M Hingorani
- Molecular Biology and Biochemistry Department, ‡Molecular Biophysics Program, §Chemistry Department, and ∥Computer Science Department, Wesleyan University , Middletown, Connecticut 06459, United States
| | - David L Beveridge
- Molecular Biology and Biochemistry Department, ‡Molecular Biophysics Program, §Chemistry Department, and ∥Computer Science Department, Wesleyan University , Middletown, Connecticut 06459, United States
| |
Collapse
|
26
|
Tian Z, Liu J, Zhang Y. Key Residues Involved in the Interaction between Cydia pomonella Pheromone Binding Protein 1 (CpomPBP1) and Codlemone. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7994-8001. [PMID: 27709920 DOI: 10.1021/acs.jafc.6b02843] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Codlemone exhibited high affinity to CpomPBP1; studying their binding mode can provide insights into the rational design of active semiochemicals. Our findings suggested that residues including Phe12, Phe36, Trp37, Ile52, Ile 94, Ala115, and Phe118 were favorable to the binding of codlemone to CpomPBP1, whereas residues providing unfavorable contributions such as Ser56 were negative to the binding. van der Waals energy and electrostatic energy, mainly derived from the side chains of favorable residues, contributed most to the formation and stability of the CpomPBP1-codlemone complex. Of the residues involved in the interaction between CpomPBP1 and codlemone, Phe12 and Trp37, the mutation of which into Ala caused a significant decrease of CpomPBP1 binding ability, were two key residues in determining the binding affinity of codlemone to CpomPBP1. This study shed light on discovering novel active semiochemicals as well as facilitating chemical modification of lead semiochemicals.
Collapse
Affiliation(s)
- Zhen Tian
- Key Laboratory of Plant Protection Resources & Pest Management of the Ministry of Education, College of Plant Protection, Northwest A&F University , Yangling 712100, Shaanxi, China
| | - Jiyuan Liu
- Key Laboratory of Plant Protection Resources & Pest Management of the Ministry of Education, College of Plant Protection, Northwest A&F University , Yangling 712100, Shaanxi, China
| | - Yalin Zhang
- Key Laboratory of Plant Protection Resources & Pest Management of the Ministry of Education, College of Plant Protection, Northwest A&F University , Yangling 712100, Shaanxi, China
| |
Collapse
|
27
|
Wang YT, Cheng TL. Refined models of New Delhi metallo-beta-lactamase-1 with inhibitors: an QM/MM modeling study. J Biomol Struct Dyn 2016; 34:2214-23. [DOI: 10.1080/07391102.2015.1110834] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Yeng-Tseng Wang
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan, P.R. China
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan, P.R. China
| | - Tian-Lu Cheng
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan, P.R. China
| |
Collapse
|
28
|
Enhancing the thermal tolerance of a cis-epoxysuccinate hydrolase via combining directed evolution with various semi-rational redesign methods. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.molcatb.2015.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Munteanu CR, Pimenta AC, Fernandez-Lozano C, Melo A, Cordeiro MNDS, Moreira IS. Solvent accessible surface area-based hot-spot detection methods for protein-protein and protein-nucleic acid interfaces. J Chem Inf Model 2015; 55:1077-86. [PMID: 25845030 DOI: 10.1021/ci500760m] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Due to the importance of hot-spots (HS) detection and the efficiency of computational methodologies, several HS detecting approaches have been developed. The current paper presents new models to predict HS for protein-protein and protein-nucleic acid interactions with better statistics compared with the ones currently reported in literature. These models are based on solvent accessible surface area (SASA) and genetic conservation features subjected to simple Bayes networks (protein-protein systems) and a more complex multi-objective genetic algorithm-support vector machine algorithms (protein-nucleic acid systems). The best models for these interactions have been implemented in two free Web tools.
Collapse
Affiliation(s)
- Cristian R Munteanu
- †Information and Communication Technologies Department, Computer Science Faculty, University of A Coruna, Campus de Elviña s/n, 15071 A Coruña, Spain
| | - António C Pimenta
- ‡REQUIMTE/Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Carlos Fernandez-Lozano
- †Information and Communication Technologies Department, Computer Science Faculty, University of A Coruna, Campus de Elviña s/n, 15071 A Coruña, Spain
| | - André Melo
- ‡REQUIMTE/Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Maria N D S Cordeiro
- ‡REQUIMTE/Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Irina S Moreira
- ‡REQUIMTE/Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal.,§CNC-Center for Neuroscience and Cell Biology, Universidade de Coimbra, Rua Larga, FMUC, Polo I, 1°andar, 3004-517 Coimbra, Portugal
| |
Collapse
|