1
|
Wu Q, Xiao Y, Yang X, Zhu A, Cao W, Cai L, Lin X, Zhao Z, Zhang Q, Zhou X. Magnetic-assisted and aptamer-based SERS biosensor for high enrichment, ultrasensitive detection of multicomponent heart failure biomarkers. Talanta 2025; 290:127834. [PMID: 40020612 DOI: 10.1016/j.talanta.2025.127834] [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: 01/10/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
The high-sensitivity detection of low-concentration multicomponent biomarkers in the blood of heart failure (HF) patients using surface-enhanced Raman spectroscopy (SERS) remains a significant challenge. In this study, an ultrasensitive biosensor for the detection of multicomponent HF biomarkers was designed. This biosensor utilizes Au@Ag nanoparticles (Au@Ag NPs) functionalized with Raman reporter molecules (RaRs) as SERS probes, and Ag-coated Fe3O4 nanoparticles (Fe3O4-Ag NPs) modified with internal standard (IS) molecules as the capture substrate, offering the dual advantages of magnetic enrichment and SERS enhancement. Additionally, specific aptamers or antibodies were conjugated to the surfaces of Au@Ag NPs and Fe3O4-Ag NPs to specifically recognize target proteins to construct a three-layer composite structure (Fe3O4-Ag/HF biomarkers/Au@Ag). The limit of detection (LOD) of HF markers for cTnI, NT-proBNP, and sST2 is 0.1 pg/mL, 1.0 fg/mL, and 1.0 fg/mL, respectively, surpassing most reported methods. Additionally, the analysis of 45 clinical serum samples revealed no statistically significant differences between the SERS-based results and those obtained by conventional clinical methods, as confirmed by the Shapiro-Wilk test (p > 0.05). In conclusion, this SERS biosensor successfully developed an easy-to-operate accurate diagnosis method capable of simultaneous, quantitative detection of multiple HF biomarkers and provided a new technique for accurate diagnosis of other diseases in clinical testing.
Collapse
Affiliation(s)
- Qingyu Wu
- Department of Pharmacy, Shantou University Medical College, Shantou, Guangdong, 515041, China; Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China; College of Medical Technology, Zhangzhou Health Vocational College, Zhangzhou, Fujian, 363000, China
| | - Yingxiu Xiao
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Xinran Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Aoxue Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Wendi Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Leshan Cai
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Xiaozhe Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Zhenhua Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Qiaoxin Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China.
| | - Xia Zhou
- Department of Pharmacy, Shantou University Medical College, Shantou, Guangdong, 515041, China.
| |
Collapse
|
2
|
Dumas N, Portelli G, Ji Y, Dupont F, Jendoubi M, Lalli E. Detection of protein structural hotspots using AI distillation and explainability: application to the DAX-1 protein. NAR Genom Bioinform 2025; 7:lqaf047. [PMID: 40264682 PMCID: PMC12012785 DOI: 10.1093/nargab/lqaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/26/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025] Open
Abstract
AlphaMissense is a valuable resource for discerning important functional regions within proteins, providing pathogenicity heatmaps that highlight the pathogenic risk of specific mutations along the protein sequence. However, due to protein folding and long-range interactions, the actual structural alterations with functional implications may be occurring at a distance from the mutation site. As a result, the identification of the most sensitive structural regions for protein function may be hampered by the presence of mutations that indirectly affect the critical regions from a distance. In this study, we illustrate how the use of AlphaMissense predictions to train an XGBoost regression model on structural features extracted from the structures of protein variants predicted by OmegaFold enables the definition of a new explainability metric: a residue-based importance score that highlights the most critical structural domains within a protein sequence. To verify the accuracy of our approach, we applied it to the extensively studied protein DAX-1 and successfully identified critical structural domains. Notably, as this score only requires knowledge of the protein's amino acid sequence, it is valuable in guiding experimental investigations aimed at discovering functionally crucial regions in proteins that have been poorly characterized.
Collapse
Affiliation(s)
- Noé Dumas
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Geoffrey Portelli
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Yang Ji
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Florent Dupont
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Mehdi Jendoubi
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Enzo Lalli
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne—Sophia Antipolis, France
- Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne—Sophia Antipolis, France
- Université Côte d’Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne—Sophia Antipolis, France
| |
Collapse
|
3
|
Chen YC, Sargsyan K, Wright JD, Chen YH, Huang YS, Lim C. PPI-hotspot ID for detecting protein-protein interaction hot spots from the free protein structure. eLife 2024; 13:RP96643. [PMID: 39283314 PMCID: PMC11405013 DOI: 10.7554/elife.96643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Experimental detection of residues critical for protein-protein interactions (PPI) is a time-consuming, costly, and labor-intensive process. Hence, high-throughput PPI-hot spot prediction methods have been developed, but they have been validated using relatively small datasets, which may compromise their predictive reliability. Here, we introduce PPI-hotspotID, a novel method for identifying PPI-hot spots using the free protein structure, and validated it on the largest collection of experimentally confirmed PPI-hot spots to date. We explored the possibility of detecting PPI-hot spots using (i) FTMap in the PPI mode, which identifies hot spots on protein-protein interfaces from the free protein structure, and (ii) the interface residues predicted by AlphaFold-Multimer. PPI-hotspotID yielded better performance than FTMap and SPOTONE, a webserver for predicting PPI-hot spots given the protein sequence. When combined with the AlphaFold-Multimer-predicted interface residues, PPI-hotspotID yielded better performance than either method alone. Furthermore, we experimentally verified several PPI-hotspotID-predicted PPI-hot spots of eukaryotic elongation factor 2. Notably, PPI-hotspotID can reveal PPI-hot spots not obvious from complex structures, including those in indirect contact with binding partners. PPI-hotspotID serves as a valuable tool for understanding PPI mechanisms and aiding drug design. It is available as a web server (https://ppihotspotid.limlab.dnsalias.org/) and open-source code (https://github.com/wrigjz/ppihotspotid/).
Collapse
Affiliation(s)
- Yao Chi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Karen Sargsyan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jon D Wright
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Hsien Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Carmay Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
4
|
Janke JJ, Starr CG, Kingsbury JS, Furtmann N, Roberts CJ, Calero-Rubio C. Computational Screening for mAb Colloidal Stability with Coarse-Grained, Molecular-Scale Simulations. J Phys Chem B 2024; 128:1515-1526. [PMID: 38315822 DOI: 10.1021/acs.jpcb.3c05303] [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: 02/07/2024]
Abstract
Monoclonal antibodies (mAbs) are an important modality of protein therapeutics with broad applications for numerous diseases. However, colloidal instabilities occurring at high protein concentrations can limit the ability to develop stable, high-concentration liquid dosage forms that are required for patient-centric, device-mediated products. Therefore, it is advantageous to identify colloidally stable mAbs early in the discovery process to ensure that they are selected for development. Experimental screening for colloidal stability can be time- and resource-consuming and is most feasible at the later stages of drug development due to material requirements. Alternatively, computational approaches have emerging potential to provide efficient screening and focus developmental efforts on mAbs with the greatest developability potential, while providing mechanistic relationships for colloidal instability. In this work, coarse-grained, molecular-scale models were fine-tuned to screen for colloidal stability at amino-acid resolution. This model parameterization provides a framework to screen for mAb self-interactions and extrapolate to bulk solution behavior. This approach was applied to a wide array of mAbs under multiple buffer conditions, demonstrating the utility of the presented computational approach to augment early candidate screening and later formulation strategies for protein therapeutics.
Collapse
Affiliation(s)
- J Joel Janke
- Biologics Drug Product Development and Manufacturing, Sanofi, Framingham, Massachusetts 01701, United States
| | - Charles G Starr
- Biologics Drug Product Development and Manufacturing, Sanofi, Framingham, Massachusetts 01701, United States
| | - Jonathan S Kingsbury
- Biologics Drug Product Development and Manufacturing, Sanofi, Framingham, Massachusetts 01701, United States
| | - Norbert Furtmann
- Large Molecules Research Platform, Sanofi-Aventis Deutschland GmbH, Frankfurt 65926, Germany
| | - Christopher J Roberts
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Cesar Calero-Rubio
- Biologics Drug Product Development and Manufacturing, Sanofi, Framingham, Massachusetts 01701, United States
| |
Collapse
|
5
|
Jarończyk M, Abagyan R, Totrov M. Software and Databases for Protein-Protein Docking. Methods Mol Biol 2024; 2780:129-138. [PMID: 38987467 DOI: 10.1007/978-1-0716-3985-6_8] [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] [Indexed: 07/12/2024]
Abstract
Protein-protein interactions (PPIs) provide valuable insights for understanding the principles of biological systems and for elucidating causes of incurable diseases. One of the techniques used for computational prediction of PPIs is protein-protein docking calculations, and a variety of software has been developed. This chapter is a summary of software and databases used for protein-protein docking.
Collapse
Affiliation(s)
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
6
|
Wu M, Luan J, Zhang D, Fan H, Qiao L, Zhang C. Development and validation of a clinical prediction model for glioma grade using machine learning. Technol Health Care 2024; 32:1977-1990. [PMID: 38306068 DOI: 10.3233/thc-231645] [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] [Indexed: 02/03/2024]
Abstract
BACKGROUND Histopathological evaluation is currently the gold standard for grading gliomas; however, this technique is invasive. OBJECTIVE This study aimed to develop and validate a diagnostic prediction model for glioma by employing multiple machine learning algorithms to identify risk factors associated with high-grade glioma, facilitating the prediction of glioma grading. METHODS Data from 1114 eligible glioma patients were obtained from The Cancer Genome Atlas (TCGA) database, which was divided into a training set (n= 781) and a test set (n= 333). Fifty machine learning algorithms were employed, and the optimal algorithm was selected to construct a prediction model. The performance of the machine learning prediction model was compared to the clinical prediction model in terms of discrimination, calibration, and clinical validity to assess the performance of the prediction model. RESULTS The area under the curve (AUC) values of the machine learning prediction models (training set: 0.870 vs. 0.740, test set: 0.863 vs. 0.718) were significantly improved from the clinical prediction models. Furthermore, significant improvement in discrimination was observed for the Integrated Discrimination Improvement (IDI) (training set: 0.230, test set: 0.270) and Net Reclassification Index (NRI) (training set: 0.170, test set: 0.170) from the clinical prognostic model. Both models showed a high goodness of fit and an increased net benefit. CONCLUSION A strong prediction accuracy model can be developed using machine learning algorithms to screen for high-grade glioma risk predictors, which can serve as a non-invasive prediction tool for preoperative diagnostic grading of glioma.
Collapse
Affiliation(s)
- Mingzhen Wu
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China
| | - Jixin Luan
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China
| | - Di Zhang
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China
| | - Hua Fan
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China
| | - Lishan Qiao
- School of Mathematics, Liaocheng University, Shandong, China
| | - Chuanchen Zhang
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China
| |
Collapse
|
7
|
Ntallis C, Tzoupis H, Tselios T, Chasapis CT, Vlamis-Gardikas A. Distinct or Overlapping Areas of Mitochondrial Thioredoxin 2 May Be Used for Its Covalent and Strong Non-Covalent Interactions with Protein Ligands. Antioxidants (Basel) 2023; 13:15. [PMID: 38275635 PMCID: PMC10812433 DOI: 10.3390/antiox13010015] [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: 11/01/2023] [Revised: 12/09/2023] [Accepted: 12/16/2023] [Indexed: 01/27/2024] Open
Abstract
In silico approaches were employed to examine the characteristics of interactions between human mitochondrial thioredoxin 2 (HsTrx2) and its 38 previously identified mitochondrial protein ligands. All interactions appeared driven mainly by electrostatic forces. The statistically significant residues of HsTrx2 for interactions were characterized as "contact hot spots". Since these were identical/adjacent to putative thermodynamic hot spots, an energy network approach identified their neighbors to highlight possible contact interfaces. Three distinct areas for binding emerged: (i) one around the active site for covalent interactions, (ii) another antipodal to the active site for strong non-covalent interactions, and (iii) a third area involved in both kinds of interactions. The contact interfaces of HsTrx2 were projected as respective interfaces for Escherichia coli Trx1 (EcoTrx1), 2, and HsTrx1. Comparison of the interfaces and contact hot spots of HsTrx2 to the contact residues of EcoTx1 and HsTrx1 from existing crystal complexes with protein ligands supported the hypothesis, except for a part of the cleft/groove adjacent to Trp30 preceding the active site. The outcomes of this study raise the possibility for the rational design of selective inhibitors for the interactions of HsTrx2 with specific protein ligands without affecting the entirety of the functions of the Trx system.
Collapse
Affiliation(s)
- Charalampos Ntallis
- Department of Chemistry, University of Patras, 26504 Rion, Greece; (C.N.); (H.T.); (T.T.)
| | - Haralampos Tzoupis
- Department of Chemistry, University of Patras, 26504 Rion, Greece; (C.N.); (H.T.); (T.T.)
| | - Theodore Tselios
- Department of Chemistry, University of Patras, 26504 Rion, Greece; (C.N.); (H.T.); (T.T.)
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, Vas. Constantinou 48, 11635 Athens, Greece;
| | | |
Collapse
|
8
|
Ahsan T, Shoily SS, Ahmed T, Sajib AA. Role of the redox state of the Pirin-bound cofactor on interaction with the master regulators of inflammation and other pathways. PLoS One 2023; 18:e0289158. [PMID: 38033031 PMCID: PMC10688961 DOI: 10.1371/journal.pone.0289158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/10/2023] [Indexed: 12/02/2023] Open
Abstract
Persistent cellular stress induced perpetuation and uncontrolled amplification of inflammatory response results in a shift from tissue repair toward collateral damage, significant alterations of tissue functions, and derangements of homeostasis which in turn can lead to a large number of acute and chronic pathological conditions, such as chronic heart failure, atherosclerosis, myocardial infarction, neurodegenerative diseases, diabetes, rheumatoid arthritis, and cancer. Keeping the vital role of balanced inflammation in maintaining tissue integrity in mind, the way to combating inflammatory diseases may be through identification and characterization of mediators of inflammation that can be targeted without hampering normal body function. Pirin (PIR) is a non-heme iron containing protein having two different conformations depending on the oxidation state of the iron. Through exploration of the Pirin interactome and using molecular docking approaches, we identified that the Fe2+-bound Pirin directly interacts with BCL3, NFKBIA, NFIX and SMAD9 with more resemblance to the native binding pose and higher affinity than the Fe3+-bound form. In addition, Pirin appears to have a function in the regulation of inflammation, the transition between the canonical and non-canonical NF-κB pathways, and the remodeling of the actin cytoskeleton. Moreover, Pirin signaling appears to have a critical role in tumor invasion and metastasis, as well as metabolic and neuro-pathological complications. There are regulatory variants in PIR that can influence expression of not only PIR but also other genes, including VEGFD and ACE2. Disparity exists between South Asian and European populations in the frequencies of variant alleles at some of these regulatory loci that may lead to differential occurrence of Pirin-mediated pathogenic conditions.
Collapse
Affiliation(s)
- Tamim Ahsan
- Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka, Bangladesh
| | - Sabrina Samad Shoily
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Tasnim Ahmed
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
9
|
Yadav AK, Nagar BC, Pradhan G. FPGA Implementation of IIR Notch and Anti-Notch Filters With an Application to Localization of Protein Hot-Spots. IEEE Trans Nanobioscience 2023; 22:863-871. [PMID: 37022064 DOI: 10.1109/tnb.2023.3238733] [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: 01/25/2023]
Abstract
In this paper, high-speed second-order infinite impulse response (IIR) notch filter (NF) and anti-notch filter (ANF) are designed and realized on hardware. The improvement in speed of operation for the NF is then achieved by using the re-timing concept. The ANF is designed to specify a stability margin and minimize the amplitude area. Next, an improved approach is proposed for the detection of protein hot-spot locations using the designed second-order IIR ANF. The analytical and experimental results reported in this paper show that the proposed approach provides better hot-spot prediction compared to the reported classical filtering techniques based on the IIR Chebyshev filter and S-transform. The proposed approach also yields consistency in prediction hot-spots compared to the results based on biological methodologies. Furthermore, the presented technique reveals some new "potential" hot-spots. The proposed filters are simulated and synthesized using the Xilinx Vivado 18.3 software platform with Zynq-7000 Series (ZedBoard Zynq Evaluation and Development Kit xc7z020clg484-1) FPGA family.
Collapse
|
10
|
Zhang Y, Yao S, Chen P. Prediction of hot spots towards drug discovery by protein sequence embedding with 1D convolutional neural network. PLoS One 2023; 18:e0290899. [PMID: 37721924 PMCID: PMC10506709 DOI: 10.1371/journal.pone.0290899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Protein hotspot residues are key sites that mediate protein-protein interactions. Accurate identification of these residues is essential for understanding the mechanism from protein to function and for designing drug targets. Current research has mostly focused on using machine learning methods to predict hot spots from known interface residues, which artificially extract the corresponding features of amino acid residues from sequence, structure, evolution, energy, and other information to train and test machine learning models. The process is cumbersome, time-consuming and laborious to some extent. This paper proposes a novel idea that develops a pre-trained protein sequence embedding model combined with a one-dimensional convolutional neural network, called Embed-1dCNN, to predict protein hotspot residues. In order to obtain large data samples, this work integrates and extracts data from the datasets of ASEdb, BID, SKEMPI and dbMPIKT to generate a new dataset, and adopts the SMOTE algorithm to expand positive samples to form the training set. The experimental results show that the method achieves an F1 score of 0.82 on the test set. Compared with other hot spot prediction methods, our model achieved better prediction performance.
Collapse
Affiliation(s)
- Youzhi Zhang
- School of Computer and Information, Anqing Normal University, Anqing, China
- University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing, China
- National Engineering Research Center for Agro-Ecological Big Data Analysis & Application, Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology & School of Internet, Anhui University, Anhui, China
| | - Sijie Yao
- National Engineering Research Center for Agro-Ecological Big Data Analysis & Application, Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology & School of Internet, Anhui University, Anhui, China
| | - Peng Chen
- School of Computer and Information, Anqing Normal University, Anqing, China
- National Engineering Research Center for Agro-Ecological Big Data Analysis & Application, Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology & School of Internet, Anhui University, Anhui, China
| |
Collapse
|
11
|
Pal S, Mehta P, Pandey A, Ara A, Ghoshal U, Ghoshal UC, Pandey R, Tripathi RK, Yadav PN, Ravishankar R, Kundu TK, Rajender S. Molecular determinants associated with temporal succession of SARS-CoV-2 variants in Uttar Pradesh, India. Front Microbiol 2023; 14:986729. [PMID: 36819024 PMCID: PMC9929466 DOI: 10.3389/fmicb.2023.986729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
The emergence and rapid evolution of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) caused a global crisis that required a detailed characterization of the dynamics of mutational pattern of the viral genome for comprehending its epidemiology, pathogenesis and containment. We investigated the molecular evolution of the SASR-CoV-2 genome during the first, second and third waves of COVID-19 in Uttar Pradesh, India. Nanopore sequencing of the SARS-CoV-2 genome was undertaken in 544 confirmed cases of COVID-19, which included vaccinated and unvaccinated individuals. In the first wave (unvaccinated population), the 20A clade (56.32%) was superior that was replaced by 21A Delta in the second wave, which was more often seen in vaccinated individuals in comparison to unvaccinated (75.84% versus 16.17%, respectively). Subsequently, 21A delta got outcompeted by Omicron (71.8%), especially the 21L variant, in the third wave. We noticed that Q677H appeared in 20A Alpha and stayed up to Delta, D614G appeared in 20A Alpha and stayed in Delta and Omicron variants (got fixed), and several other mutations appeared in Delta and stayed in Omicron. A cross-sectional analysis of the vaccinated and unvaccinated individuals during the second wave revealed signature combinations of E156G, F157Del, L452R, T478K, D614G mutations in the Spike protein that might have facilitated vaccination breach in India. Interestingly, some of these mutation combinations were carried forward from Delta to Omicron. In silico protein docking showed that Omicron had a higher binding affinity with the host ACE2 receptor, resulting in enhanced infectivity of Omicron over the Delta variant. This work has identified the combinations of key mutations causing vaccination breach in India and provided insights into the change of [virus's] binding affinity with evolution, resulting in more virulence in Delta and more infectivity in Omicron variants of SARS-CoV-2. Our findings will help in understanding the COVID-19 disease biology and guide further surveillance of the SARS-CoV-2 genome to facilitate the development of vaccines with better efficacies.
Collapse
Affiliation(s)
- Smita Pal
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India
| | - Poonam Mehta
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ankita Pandey
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Anam Ara
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India
| | - Ujjala Ghoshal
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Uday C. Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Rajesh Pandey
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Raj Kamal Tripathi
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Prem N. Yadav
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ramachandran Ravishankar
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Tapas K. Kundu
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Singh Rajender
- CSIR-Central Drug Research Institute, Lucknow (CSIR-CDRI), Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,*Correspondence: Singh Rajender, ✉
| |
Collapse
|
12
|
Tiwari V, Sowdhamini R. Structural modelling and dynamics of full-length of TLR10 sheds light on possible modes of dimerization, ligand binding and mechanism of action. Curr Res Struct Biol 2023; 5:100097. [PMID: 36911652 PMCID: PMC9996232 DOI: 10.1016/j.crstbi.2023.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 02/01/2023] [Accepted: 02/05/2023] [Indexed: 02/22/2023] Open
Abstract
Toll like receptors (TLRs) play a pivotal role in innate and adaptive immunity. There are 10 TLRs in the human genome, of which TLR10 is the least characterized. Genetic polymorphism of TLR10 has been shown to be associated with multiple diseases including tuberculosis and rheumatoid arthritis. TLR10 consists of an extracellular domain (ECD), a single-pass transmembrane (TM) helix and intracellular TIR (Toll/Interleukin-1 receptor) domain. ECD is employed for ligand recognition and the intracellular domain interacts with other TIR domain-containing adapter proteins for signal transduction. Experimental structure of ECD or TM domain is not available for TLR10. In this study, we have modelled multiple forms of TLR10-ECD dimers, such as closed and open forms, starting from available structures of homologues. Subsequently, multiple full-length TLR10 homodimer models were generated by utilizing homology modelling and protein-protein docking. The dynamics of these models in membrane-aqueous environment revealed the global motion of ECD and TIR domain towards membrane bilayer. The TIR domain residues exhibited high root mean square fluctuation compared to ECD. The 'closed form' model was observed to be energetically more favorable than 'open form' model. The evaluation of persistent interchain interactions, along with their conservation score, unveiled critical residues for each model. Further, the binding of dsRNA to TLR10 was modelled by defined and blind docking approaches. Differential binding of dsRNA to the protomers of TLR10 was observed upon simulation that could provide clues on ligand disassociation. Dynamic network analysis revealed that the 'open form' model can be the functional form while 'closed form' model can be the apo form of TLR10.
Collapse
Affiliation(s)
- Vikas Tiwari
- National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - R Sowdhamini
- National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| |
Collapse
|
13
|
Scietti L, Forneris F. Modeling of Protein Complexes. Methods Mol Biol 2023; 2627:349-371. [PMID: 36959458 DOI: 10.1007/978-1-0716-2974-1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
The recent advances in structural biology, combined with continuously increasing computational capabilities and development of advanced softwares, have drastically simplified the workflow for protein homology modeling. Modeling of individual proteins is nowadays quick and straightforward for a large variety of protein targets, thanks to guided pipelines relying on advanced computational tools and user-friendly interfaces, which have extended and promoted the use of modeling also to scientists not focusing on molecular structures of proteins. Nevertheless, construction of models of multi-protein complexes remains quite challenging for the non-experts, often due to the usage of specific procedures depending on the system under investigation and the need for experimental validation approaches to strengthen the generated output.In this chapter, we provide a brief overview of the approaches enabling generation of multi-protein complex models starting from homology models of individual protein components. Using real-life examples, we include two examples to guide the reader in the generation of homomeric and heteromeric protein models.
Collapse
Affiliation(s)
- Luigi Scietti
- Department of Biology and Biotechnology, The Armenise-Harvard Laboratory of Structural Biology, University of Pavia, Pavia, Italy.
| | - Federico Forneris
- Department of Biology and Biotechnology, The Armenise-Harvard Laboratory of Structural Biology, University of Pavia, Pavia, Italy.
| |
Collapse
|
14
|
Shirzadeh M, Monhemi H, Eftekhari M. Large interfacial relocation in RBD-ACE2 complex may explain fast-spreading property of Omicron. J Mol Struct 2022; 1270:133842. [PMID: 35937157 PMCID: PMC9339243 DOI: 10.1016/j.molstruc.2022.133842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/07/2022] [Accepted: 07/31/2022] [Indexed: 02/04/2023]
Abstract
The Omicron variant of SARS-CoV-2 emerged in South African in late 2021. This variant has a large number of mutations, and regarded as fastest-spreading Covid variant. The spike RBD region of SARS-CoV-2 and its interaction with human ACE2 play fundamental role in viral infection and transmission. To explore the reason of fast-spreading properties of Omicron variant, we have modeled the interactions of Omicron RBD and human ACE2 using docking and molecular dynamics simulations. Results show that RBD-ACE2 binding site may drastically relocate with an enlarged interface. The predicted interface has large negative binding energies and shows stable conformation in molecular dynamics simulations. It was found that the interfacial area in Omicron RBD-ACE2 complex is increased up to 40% in comparison to wild-type Sars-Cov-2. Moreover, the number of hydrogen bonds significantly increased up to 80%. The key interacting residues become also very different in Omicron variant. The new binding interface can significantly accommodate R403, as a key RBD residue, near ACE2 surface which leads to two new strong salt bridges. The exploration of the new binding interface can help to understand the reasons of high transmission rate of Omicron.
Collapse
Affiliation(s)
- Maryam Shirzadeh
- Departemant of Chemistry, Faculty of Science, University of Neyshabur
| | - Hassan Monhemi
- Departemant of Chemistry, Faculty of Science, University of Neyshabur
| | | |
Collapse
|
15
|
Wu C, Guo D. Computational Docking Reveals Co-Evolution of C4 Carbon Delivery Enzymes in Diverse Plants. Int J Mol Sci 2022; 23:12688. [PMID: 36293547 PMCID: PMC9604239 DOI: 10.3390/ijms232012688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
Proteins are modular functionalities regulating multiple cellular activities in prokaryotes and eukaryotes. As a consequence of higher plants adapting to arid and thermal conditions, C4 photosynthesis is the carbon fixation process involving multi-enzymes working in a coordinated fashion. However, how these enzymes interact with each other and whether they co-evolve in parallel to maintain interactions in different plants remain elusive to date. Here, we report our findings on the global protein co-evolution relationship and local dynamics of co-varying site shifts in key C4 photosynthetic enzymes. We found that in most of the selected key C4 photosynthetic enzymes, global pairwise co-evolution events exist to form functional couplings. Besides, protein-protein interactions between these enzymes may suggest their unknown functionalities in the carbon delivery process. For PEPC and PPCK regulation pairs, pocket formation at the interactive interface are not necessary for their function. This feature is distinct from another well-known regulation pair in C4 photosynthesis, namely, PPDK and PPDK-RP, where the pockets are necessary. Our findings facilitate the discovery of novel protein regulation types and contribute to expanding our knowledge about C4 photosynthesis.
Collapse
Affiliation(s)
| | - Dianjing Guo
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
16
|
Kitel R, Rodríguez I, del Corte X, Atmaj J, Żarnik M, Surmiak E, Muszak D, Magiera-Mularz K, Popowicz GM, Holak TA, Musielak B. Exploring the Surface of the Ectodomain of the PD-L1 Immune Checkpoint with Small-Molecule Fragments. ACS Chem Biol 2022; 17:2655-2663. [PMID: 36073782 PMCID: PMC9486809 DOI: 10.1021/acschembio.2c00583] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Development of small molecules targeting the PD-L1/PD-1 interface is advancing both in industry and academia, but only a few have reached early-stage clinical trials. Here, we take a closer look at the general druggability of PD-L1 using in silico hot spot mapping and nuclear magnetic resonance (NMR)-based characterization. We found that the conformational elasticity of the PD-L1 surface strongly influences the formation of hot spots. We deconstructed several generations of known inhibitors into fragments and examined their binding properties using differential scanning fluorimetry (DSF) and protein-based nuclear magnetic resonance (NMR). These biophysical analyses showed that not all fragments bind to the PD-L1 ectodomain despite having the biphenyl scaffold. Although most of the binding fragments induced PD-L1 oligomerization, two compounds, TAH35 and TAH36, retain the monomeric state of proteins upon binding. Additionally, the presence of the entire ectodomain did not affect the binding of the hit compounds and dimerization of PD-L1. The data demonstrated here provide important information on the PD-L1 druggability and the structure-activity relationship of the biphenyl core moiety and therefore may aid in the design of novel inhibitors and focused fragment libraries for PD-L1.
Collapse
Affiliation(s)
- Radoslaw Kitel
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Ismael Rodríguez
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Xabier del Corte
- Departamento
de Química Orgánica I, Centro de Investigación
y Estudios Avanzados “Lucio Lascaray” − Facultad
de Farmacia, University of the Basque Country, UPV/EHU Paseo de la Universidad
7, 01006 Vitoria-Gasteiz, Spain
| | - Jack Atmaj
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Magdalena Żarnik
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Ewa Surmiak
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Damian Muszak
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Katarzyna Magiera-Mularz
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Grzegorz M. Popowicz
- Institute
of Structural Biology, Helmholtz Zentrum
München, Ingolstädter
Landstrasse 1, 85764 Neuherberg, Germany
| | - Tad A. Holak
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Bogdan Musielak
- Faculty
of Chemistry, Organic Chemistry Department, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland,
| |
Collapse
|
17
|
Santa-Coloma TA. Overlapping synthetic peptides as a tool to map protein-protein interactions ̶ FSH as a model system of nonadditive interactions. Biochim Biophys Acta Gen Subj 2022; 1866:130153. [DOI: 10.1016/j.bbagen.2022.130153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
|
18
|
Chen YC, Chen YH, Wright JD, Lim C. PPI-Hotspot DB: Database of Protein-Protein Interaction Hot Spots. J Chem Inf Model 2022; 62:1052-1060. [PMID: 35147037 DOI: 10.1021/acs.jcim.2c00025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Single-point mutations of certain residues (so-called hot spots) impair/disrupt protein-protein interactions (PPIs), leading to pathogenesis and drug resistance. Conventionally, a PPI-hot spot is identified when its replacement decreased the binding free energy significantly, generally by ≥2 kcal/mol. The relatively few mutations with such a significant binding free energy drop limited the number of distinct PPI-hot spots. By defining PPI-hot spots based on mutations that have been manually curated in UniProtKB to significantly impair/disrupt PPIs in addition to binding free energy changes, we have greatly expanded the number of distinct PPI-hot spots by an order of magnitude. These experimentally determined PPI-hot spots along with available structures have been collected in a database called PPI-HotspotDB. We have applied the PPI-HotspotDB to create a nonredundant benchmark, PPI-Hotspot+PDBBM, for assessing methods to predict PPI-hot spots using the free structure as input. PPI-HotspotDB will benefit the design of mutagenesis experiments and development of PPI-hot spot prediction methods. The database and benchmark are freely available at https://ppihotspot.limlab.dnsalias.org.
Collapse
Affiliation(s)
- Yao Chi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yu-Hsien Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Jon D Wright
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Carmay Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
19
|
Big data and artificial intelligence (AI) methodologies for computer-aided drug design (CADD). Biochem Soc Trans 2022; 50:241-252. [PMID: 35076690 PMCID: PMC9022974 DOI: 10.1042/bst20211240] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022]
Abstract
There have been numerous advances in the development of computational and statistical methods and applications of big data and artificial intelligence (AI) techniques for computer-aided drug design (CADD). Drug design is a costly and laborious process considering the biological complexity of diseases. To effectively and efficiently design and develop a new drug, CADD can be used to apply cutting-edge techniques to various limitations in the drug design field. Data pre-processing approaches, which clean the raw data for consistent and reproducible applications of big data and AI methods are introduced. We include the current status of the applicability of big data and AI methods to drug design areas such as the identification of binding sites in target proteins, structure-based virtual screening (SBVS), and absorption, distribution, metabolism, excretion and toxicity (ADMET) property prediction. Data pre-processing and applications of big data and AI methods enable the accurate and comprehensive analysis of massive biomedical data and the development of predictive models in the field of drug design. Understanding and analyzing biological, chemical, or pharmaceutical architectures of biomedical entities related to drug design will provide beneficial information in the biomedical big data era.
Collapse
|
20
|
Jacobsen D, Bushara O, Mishra RK, Sun L, Liao J, Yang GY. Druggable sites/pockets of the p53-DNAJA1 protein–protein interaction: In silico modeling and in vitro/in vivo validation. Methods Enzymol 2022; 675:83-107. [DOI: 10.1016/bs.mie.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
21
|
Ovek D, Abali Z, Zeylan ME, Keskin O, Gursoy A, Tuncbag N. Artificial intelligence based methods for hot spot prediction. Curr Opin Struct Biol 2021; 72:209-218. [PMID: 34954608 DOI: 10.1016/j.sbi.2021.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/07/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022]
Abstract
Proteins interact through their interfaces to fulfill essential functions in the cell. They bind to their partners in a highly specific manner and form complexes that have a profound effect on understanding the biological pathways they are involved in. Any abnormal interactions may cause diseases. Therefore, the identification of small molecules which modulate protein interactions through their interfaces has high therapeutic potential. However, discovering such molecules is challenging. Most protein-protein binding affinity is attributed to a small set of amino acids found in protein interfaces known as hot spots. Recent studies demonstrate that drug-like small molecules specifically may bind to hot spots. Therefore, hot spot prediction is crucial. As experimental data accumulates, artificial intelligence begins to be used for computational hot spot prediction. First, we review machine learning and deep learning for computational hot spot prediction and then explain the significance of hot spots toward drug design.
Collapse
Affiliation(s)
- Damla Ovek
- College of Engineering, Koc University, 34450 Istanbul, Turkey
| | - Zeynep Abali
- College of Engineering, Koc University, 34450 Istanbul, Turkey
| | | | - Ozlem Keskin
- College of Engineering, Koc University, 34450 Istanbul, Turkey.
| | - Attila Gursoy
- College of Engineering, Koc University, 34450 Istanbul, Turkey.
| | - Nurcan Tuncbag
- College of Engineering, Koc University, 34450 Istanbul, Turkey; School of Medicine, Koc University, 34450 Istanbul, Turkey.
| |
Collapse
|
22
|
Jandova Z, Vargiu AV, Bonvin AMJJ. Native or Non-Native Protein-Protein Docking Models? Molecular Dynamics to the Rescue. J Chem Theory Comput 2021; 17:5944-5954. [PMID: 34342983 PMCID: PMC8444332 DOI: 10.1021/acs.jctc.1c00336] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Indexed: 11/29/2022]
Abstract
Molecular docking excels at creating a plethora of potential models of protein-protein complexes. To correctly distinguish the favorable, native-like models from the remaining ones remains, however, a challenge. We assessed here if a protocol based on molecular dynamics (MD) simulations would allow distinguishing native from non-native models to complement scoring functions used in docking. To this end, the first models for 25 protein-protein complexes were generated using HADDOCK. Next, MD simulations complemented with machine learning were used to discriminate between native and non-native complexes based on a combination of metrics reporting on the stability of the initial models. Native models showed higher stability in almost all measured properties, including the key ones used for scoring in the Critical Assessment of PRedicted Interaction (CAPRI) competition, namely the positional root mean square deviations and fraction of native contacts from the initial docked model. A random forest classifier was trained, reaching a 0.85 accuracy in correctly distinguishing native from non-native complexes. Reasonably modest simulation lengths of the order of 50-100 ns are sufficient to reach this accuracy, which makes this approach applicable in practice.
Collapse
Affiliation(s)
- Zuzana Jandova
- Computational
Structural Biology Group, Bijvoet Centre for Biomolecular Research,
Faculty of Science—Chemistry, Utrecht
University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Attilio Vittorio Vargiu
- Physics
Department, University of Cagliari, Cittadella
Universitaria, S.P. 8 km 0.700, 09042 Monserrato, Italy
| | - Alexandre M. J. J. Bonvin
- Computational
Structural Biology Group, Bijvoet Centre for Biomolecular Research,
Faculty of Science—Chemistry, Utrecht
University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| |
Collapse
|
23
|
Yang W, Wang K, Wu H, Shao H, Chen H, Zhu J. Peptide scaffold‐derived peptidomimetic farnesyltransferase inhibitors. J CHIN CHEM SOC-TAIP 2021. [DOI: 10.1002/jccs.202100037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Wei Yang
- Department of Infectious Diseases, Taizhou Hospital Zhejiang University Taizhou China
| | - Kuifeng Wang
- Department of Infectious Diseases, Taizhou Hospital Zhejiang University Taizhou China
| | - Hongwei Wu
- Department of Infectious Diseases Affiliated Taizhou Hospital of Wenzhou Medical University Taizhou China
| | - Hui Shao
- Department of Infectious Diseases, Taizhou Hospital Zhejiang University Taizhou China
| | - Huazhong Chen
- Department of Infectious Diseases, Taizhou Hospital Zhejiang University Taizhou China
| | - Jiansheng Zhu
- Department of Infectious Diseases, Taizhou Hospital Zhejiang University Taizhou China
| |
Collapse
|
24
|
Schoeps B, Eckfeld C, Flüter L, Keppler S, Mishra R, Knolle P, Bayerl F, Böttcher J, Hermann CD, Häußler D, Krüger A. Identification of invariant chain CD74 as a functional receptor of tissue inhibitor of metalloproteinases-1 (TIMP-1). J Biol Chem 2021; 297:101072. [PMID: 34391782 PMCID: PMC8429975 DOI: 10.1016/j.jbc.2021.101072] [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: 06/28/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 11/29/2022] Open
Abstract
Multifunctionality of tissue inhibitor of metalloproteinases-1 (TIMP-1) comprising antiproteolytic as well as cytokinic activity has been attributed to its N-terminal and C-terminal domains, respectively. The molecular basis of the emerging proinflammatory cytokinic activity of TIMP-1 is still not completely understood. The cytokine receptor invariant chain (CD74) is involved in many inflammation-associated diseases and is highly expressed by immune cells. CD74 triggers zeta chain–associated protein kinase-70 (ZAP-70) signaling–associated activation upon interaction with its only known ligand, the macrophage migration inhibitory factor. Here, we demonstrate TIMP-1–CD74 interaction by coimmunoprecipitation and confocal microscopy in cells engineered to overexpress CD74. In silico docking in HADDOCK predicted regions of the N-terminal domain of TIMP-1 (N-TIMP-1) to interact with CD74. This was experimentally confirmed by confocal microscopy demonstrating that recombinant N-TIMP-1 lacking the entire C-terminal domain was sufficient to bind CD74. Interaction of TIMP-1 with endogenously expressed CD74 was demonstrated in the Namalwa B lymphoma cell line by dot blot binding assays as well as confocal microscopy. Functionally, we demonstrated that TIMP-1–CD74 interaction triggered intracellular ZAP-70 activation. N-TIMP-1 was sufficient to induce ZAP-70 activation and interference with the cytokine-binding site of CD74 using a synthetic peptide–abrogated TIMP-1-mediated ZAP-70 activation. Altogether, we here identified CD74 as a receptor and mediator of cytokinic TIMP-1 activity and revealed TIMP-1 as moonlighting protein harboring both cytokinic and antiproteolytic activity within its N-terminal domain. Recognition of this functional TIMP-1–CD74 interaction may shed new light on clinical attempts to therapeutically target ligand-induced CD74 activity in cancer and other inflammatory diseases.
Collapse
Affiliation(s)
- Benjamin Schoeps
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Celina Eckfeld
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Laura Flüter
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Selina Keppler
- School of Medicine, Institute of Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Ritu Mishra
- School of Medicine, Institute of Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Percy Knolle
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Felix Bayerl
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Jan Böttcher
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Chris D Hermann
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Daniel Häußler
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Achim Krüger
- School of Medicine, Institutes of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany.
| |
Collapse
|
25
|
Agamennone M, Nicoli A, Bayer S, Weber V, Borro L, Gupta S, Fantacuzzi M, Di Pizio A. Protein-protein interactions at a glance: Protocols for the visualization of biomolecular interactions. Methods Cell Biol 2021; 166:271-307. [PMID: 34752337 DOI: 10.1016/bs.mcb.2021.06.012] [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] [Indexed: 01/04/2023]
Abstract
Protein-protein interactions (PPIs) play a key role in many biological processes and are intriguing targets for drug discovery campaigns. Advancements in experimental and computational techniques are leading to a growth of data accessibility, and, with it, an increased need for the analysis of PPIs. In this respect, visualization tools are essential instruments to represent and analyze biomolecular interactions. In this chapter, we reviewed some of the available tools, highlighting their features, and describing their functions with practical information on their usage.
Collapse
Affiliation(s)
| | - Alessandro Nicoli
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Sebastian Bayer
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Verena Weber
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Luca Borro
- Department of Imaging, Advanced Cardiovascular Imaging Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | | | - Antonella Di Pizio
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany.
| |
Collapse
|
26
|
Honorato RV, Koukos PI, Jiménez-García B, Tsaregorodtsev A, Verlato M, Giachetti A, Rosato A, Bonvin AMJJ. Structural Biology in the Clouds: The WeNMR-EOSC Ecosystem. Front Mol Biosci 2021; 8:729513. [PMID: 34395534 PMCID: PMC8356364 DOI: 10.3389/fmolb.2021.729513] [Citation(s) in RCA: 383] [Impact Index Per Article: 95.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/13/2021] [Indexed: 12/05/2022] Open
Abstract
Structural biology aims at characterizing the structural and dynamic properties of biological macromolecules at atomic details. Gaining insight into three dimensional structures of biomolecules and their interactions is critical for understanding the vast majority of cellular processes, with direct applications in health and food sciences. Since 2010, the WeNMR project (www.wenmr.eu) has implemented numerous web-based services to facilitate the use of advanced computational tools by researchers in the field, using the high throughput computing infrastructure provided by EGI. These services have been further developed in subsequent initiatives under H2020 projects and are now operating as Thematic Services in the European Open Science Cloud portal (www.eosc-portal.eu), sending >12 millions of jobs and using around 4,000 CPU-years per year. Here we review 10 years of successful e-infrastructure solutions serving a large worldwide community of over 23,000 users to date, providing them with user-friendly, web-based solutions that run complex workflows in structural biology. The current set of active WeNMR portals are described, together with the complex backend machinery that allows distributed computing resources to be harvested efficiently.
Collapse
Affiliation(s)
- Rodrigo V Honorato
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Department of Chemistry, Utrecht University, Utrecht, Netherlands
| | - Panagiotis I Koukos
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Department of Chemistry, Utrecht University, Utrecht, Netherlands
| | - Brian Jiménez-García
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Department of Chemistry, Utrecht University, Utrecht, Netherlands
| | | | | | - Andrea Giachetti
- Department of Chemistry and Magnetic Resonance Center, University of Florence, and C.I.R.M.M.P, Fiorentino, Italy
| | - Antonio Rosato
- Department of Chemistry and Magnetic Resonance Center, University of Florence, and C.I.R.M.M.P, Fiorentino, Italy
| | - Alexandre M J J Bonvin
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Department of Chemistry, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
27
|
Mahapatra S, Sahu SS. Integrating Resonant Recognition Model and Stockwell Transform for Localization of Hotspots in Tubulin. IEEE Trans Nanobioscience 2021; 20:345-353. [PMID: 33950844 DOI: 10.1109/tnb.2021.3077710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Tubulin is a promising target for designing anti-cancer drugs. Identification of hotspots in multifunctional Tubulin protein provides insights for new drug discovery. Although machine learning techniques have shown significant results in prediction, they fail to identify the hotspots corresponding to a particular biological function. This paper presents a signal processing technique combining resonant recognition model (RRM) and Stockwell Transform (ST) for the identification of hotspots corresponding to a particular functionality. The characteristic frequency (CF) representing a specific biological function is determined using the RRM. Then the spectrum of the protein sequence is computed using ST. The CF is filtered from the ST spectrum using a time-frequency mask. The energy peaks in the filtered sequence represent the hotspots. The hotspots predicted by the proposed method are compared with the experimentally detected binding residues of Tubulin stabilizing drug Taxol and destabilizing drug Colchicine present in the Tubulin protein. Out of the 53 experimentally identified hotspots, 60% are predicted by the proposed method whereas around 20% are predicted by existing machine learning based methods. Additionally, the proposed method predicts some new hot spots, which may be investigated.
Collapse
|
28
|
Brysbaert G, Lensink MF. Centrality Measures in Residue Interaction Networks to Highlight Amino Acids in Protein–Protein Binding. FRONTIERS IN BIOINFORMATICS 2021; 1:684970. [PMID: 36303777 PMCID: PMC9581030 DOI: 10.3389/fbinf.2021.684970] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
Residue interaction networks (RINs) describe a protein structure as a network of interacting residues. Central nodes in these networks, identified by centrality analyses, highlight those residues that play a role in the structure and function of the protein. However, little is known about the capability of such analyses to identify residues involved in the formation of macromolecular complexes. Here, we performed six different centrality measures on the RINs generated from the complexes of the SKEMPI 2 database of changes in protein–protein binding upon mutation in order to evaluate the capability of each of these measures to identify major binding residues. The analyses were performed with and without the crystallographic water molecules, in addition to the protein residues. We also investigated the use of a weight factor based on the inter-residue distances to improve the detection of these residues. We show that for the identification of major binding residues, closeness, degree, and PageRank result in good precision, whereas betweenness, eigenvector, and residue centrality analyses give a higher sensitivity. Including water in the analysis improves the sensitivity of all measures without losing precision. Applying weights only slightly raises the sensitivity of eigenvector centrality analysis. We finally show that a combination of multiple centrality analyses is the optimal approach to identify residues that play a role in protein–protein interaction.
Collapse
|
29
|
Malik RM, Fazal S, Kamal MA. Computational Analysis of Domains Vulnerable to HPV-16 E6 Oncoprotein and Corresponding Hot Spot Residues. Protein Pept Lett 2021; 28:414-425. [PMID: 32703126 DOI: 10.2174/0929866527666200722134801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/19/2020] [Accepted: 06/28/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Human Papilloma Virus (HPV) is the primary cause of cancers in cervix, head and neck regions. Oncoprotein E6 of HPV-16, after infecting human body, alters host protein- protein interaction networks. E6 interacts with several proteins, causing the infection to progress into cervical cancer. The molecular basis for these interactions is the presence of short linear peptide motifs on E6 identical to those on human proteins. METHODS Motifs of LXXLL and E/DLLL/V-G after identification on E6, were analyzed for their dynamic fluctuations by use of elastic network models. Correlation analysis of amino acid residues of E6 was also performed in specific regions of motifs. RESULTS Arginine, Leucine, Glutamine, Threonine and Glutamic acid have been identified as hot spot residues of E6 which can subsequently provide a platform for drug designing and understanding of pathogenesis of cervical cancer. These amino acids play a significant role in stabilizing interactions with host proteins, ultimately causing infections and cancers. CONCLUSION Our study validates the role of linear binding motifs of E6 of HPV in interacting with these proteins as an important event in the propagation of HPV in human cells and its transformation into cervical cancer. The study further predicts the domains of protein kinase and armadillo as part of the regions involved in the interaction of E6AP, Paxillin and TNF R1, with viral E6.
Collapse
Affiliation(s)
| | - Sahar Fazal
- Capital University of Science and Technology, Islamabad, Pakistan
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
30
|
Li G, Pahari S, Murthy AK, Liang S, Fragoza R, Yu H, Alexov E. SAAMBE-SEQ: a sequence-based method for predicting mutation effect on protein-protein binding affinity. Bioinformatics 2021; 37:992-999. [PMID: 32866236 PMCID: PMC8128451 DOI: 10.1093/bioinformatics/btaa761] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 01/04/2023] Open
Abstract
MOTIVATION Vast majority of human genetic disorders are associated with mutations that affect protein-protein interactions by altering wild-type binding affinity. Therefore, it is extremely important to assess the effect of mutations on protein-protein binding free energy to assist the development of therapeutic solutions. Currently, the most popular approaches use structural information to deliver the predictions, which precludes them to be applicable on genome-scale investigations. Indeed, with the progress of genomic sequencing, researchers are frequently dealing with assessing effect of mutations for which there is no structure available. RESULTS Here, we report a Gradient Boosting Decision Tree machine learning algorithm, the SAAMBE-SEQ, which is completely sequence-based and does not require structural information at all. SAAMBE-SEQ utilizes 80 features representing evolutionary information, sequence-based features and change of physical properties upon mutation at the mutation site. The approach is shown to achieve Pearson correlation coefficient (PCC) of 0.83 in 5-fold cross validation in a benchmarking test against experimentally determined binding free energy change (ΔΔG). Further, a blind test (no-STRUC) is compiled collecting experimental ΔΔG upon mutation for protein complexes for which structure is not available and used to benchmark SAAMBE-SEQ resulting in PCC in the range of 0.37-0.46. The accuracy of SAAMBE-SEQ method is found to be either better or comparable to most advanced structure-based methods. SAAMBE-SEQ is very fast, available as webserver and stand-alone code, and indeed utilizes only sequence information, and thus it is applicable for genome-scale investigations to study the effect of mutations on protein-protein interactions. AVAILABILITY AND IMPLEMENTATION SAAMBE-SEQ is available at http://compbio.clemson.edu/saambe_webserver/indexSEQ.php#started. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gen Li
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Swagata Pahari
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | | | - Siqi Liang
- Department of Computational Biology, Cornell University, Ithaca, NY 14850, USA
| | - Robert Fragoza
- Department of Computational Biology, Cornell University, Ithaca, NY 14850, USA
| | - Haiyuan Yu
- Department of Computational Biology, Cornell University, Ithaca, NY 14850, USA
| | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
31
|
Orr A, Wang M, Beykal B, Ganesh HS, Hearon SE, Pistikopoulos EN, Phillips TD, Tamamis P. Combining Experimental Isotherms, Minimalistic Simulations, and a Model to Understand and Predict Chemical Adsorption onto Montmorillonite Clays. ACS OMEGA 2021; 6:14090-14103. [PMID: 34124432 PMCID: PMC8190805 DOI: 10.1021/acsomega.1c00481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/11/2021] [Indexed: 05/05/2023]
Abstract
An attractive approach to minimize human and animal exposures to toxic environmental contaminants is the use of safe and effective sorbent materials to sequester them. Montmorillonite clays have been shown to tightly bind diverse toxic chemicals. Due to their promise as sorbents to mitigate chemical exposures, it is important to understand their function and rapidly screen and predict optimal clay-chemical combinations for further testing. We derived adsorption free-energy values for a structurally and physicochemically diverse set of toxic chemicals using experimental adsorption isotherms performed in the current and previous studies. We studied the diverse set of chemicals using minimalistic MD simulations and showed that their interaction energies with calcium montmorillonite clays calculated using simulation snapshots in combination with their net charge and their corresponding solvent's dielectric constant can be used as inputs to a minimalistic model to predict adsorption free energies in agreement with experiments. Additionally, experiments and computations were used to reveal structural and physicochemical properties associated with chemicals that can be adsorbed to calcium montmorillonite clay. These properties include positively charged groups, phosphine groups, halide-rich moieties, hydrogen bond donor/acceptors, and large, rigid structures. The combined experimental and computational approaches used in this study highlight the importance and potential applicability of analogous methods to study and design novel advanced sorbent systems in the future, broadening their applicability for environmental contaminants.
Collapse
Affiliation(s)
- Asuka
A. Orr
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
- Texas
A&M Energy Institute, Texas A&M
University, College
Station, Texas 77843-3372, United States
| | - Meichen Wang
- Veterinary
Integrative Biosciences Department, College of Veterinary Medicine
and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Burcu Beykal
- Texas
A&M Energy Institute, Texas A&M
University, College
Station, Texas 77843-3372, United States
| | - Hari S. Ganesh
- Texas
A&M Energy Institute, Texas A&M
University, College
Station, Texas 77843-3372, United States
| | - Sara E. Hearon
- Veterinary
Integrative Biosciences Department, College of Veterinary Medicine
and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Efstratios N. Pistikopoulos
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
- Texas
A&M Energy Institute, Texas A&M
University, College
Station, Texas 77843-3372, United States
| | - Timothy D. Phillips
- Veterinary
Integrative Biosciences Department, College of Veterinary Medicine
and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Phanourios Tamamis
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
- Texas
A&M Energy Institute, Texas A&M
University, College
Station, Texas 77843-3372, United States
- Department
of Materials Science and Engineering, Texas
A&M University, College
Station, Texas 77843-3003, United States
| |
Collapse
|
32
|
Yadav Y, Sharma SN, Shakya DK, Panchal A. Hot spots localization in proteins by optimized short time Ramanujan Fourier transform. J Bioinform Comput Biol 2021; 19:2150004. [PMID: 33819134 DOI: 10.1142/s0219720021500049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Specific functions in biological processes are dependent on protein-protein interactions. Hot spot residues play a key role in the determination of these interactions and have wide applications in engineering proteins and drug discovery. Experimental techniques to identify hotspots are often labor intensive and expensive. Also, most of the computational methods which have been developed are structure based and need some training. In this work, hotspots have been identified by sequence information alone using the Resonant Recognition Model (RRM). The proposed method uses characteristic period in place of traditionally used characteristic frequency by RRM-based methods. The characteristic period has been extracted from the consensus spectrum of protein families using the Ramanujan Fourier Transform (RFT). Position-period plots for proteins have been generated using Short Time RFT (ST-RFT) with a Gaussian window. Hot spots have been identified by thresholding of the signal corresponding to the protein's characteristic period in the ST-RFT. To enhance the performance of the ST-RFT, Gaussian window shape parameter has been optimized using concentration measure as a metric. Better sensitivity of this method has been observed compared to other reported RRM-based methods. Since the method is model independent it does not requires any training and can be readily used for any protein sequence provided its interface residues and protein family are known.
Collapse
Affiliation(s)
- Yashpal Yadav
- Department of Electronics and Instrumentation Engineering, Samrat Ashok Technological Institute, Vidisha M.P., India
| | - Sanjeev Narayan Sharma
- Department of Electronics and Instrumentation Engineering, Samrat Ashok Technological Institute, Vidisha M.P., India
| | - Devendra Kumar Shakya
- Department of Electronics and Instrumentation Engineering, Samrat Ashok Technological Institute, Vidisha M.P., India
| | - Abhishek Panchal
- Department of Electronics and Instrumentation Engineering, Samrat Ashok Technological Institute, Vidisha M.P., India
| |
Collapse
|
33
|
Matos-Filipe P, Preto AJ, Koukos PI, Mourão J, Bonvin AMJJ, Moreira IS. MENSAdb: a thorough structural analysis of membrane protein dimers. Database (Oxford) 2021; 2021:baab013. [PMID: 33822911 PMCID: PMC8023553 DOI: 10.1093/database/baab013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/19/2021] [Accepted: 03/01/2021] [Indexed: 11/14/2022]
Abstract
Membrane proteins (MPs) are key players in a variety of different cellular processes and constitute the target of around 60% of all Food and Drug Administration-approved drugs. Despite their importance, there is still a massive lack of relevant structural, biochemical and mechanistic information mainly due to their localization within the lipid bilayer. To help fulfil this gap, we developed the MEmbrane protein dimer Novel Structure Analyser database (MENSAdb). This interactive web application summarizes the evolutionary and physicochemical properties of dimeric MPs to expand the available knowledge on the fundamental principles underlying their formation. Currently, MENSAdb contains features of 167 unique MPs (63% homo- and 37% heterodimers) and brings insights into the conservation of residues, accessible solvent area descriptors, average B-factors, intermolecular contacts at 2.5 Å and 4.0 Å distance cut-offs, hydrophobic contacts, hydrogen bonds, salt bridges, π-π stacking, T-stacking and cation-π interactions. The regular update and organization of all these data into a unique platform will allow a broad community of researchers to collect and analyse a large number of features efficiently, thus facilitating their use in the development of prediction models associated with MPs. Database URL: http://www.moreiralab.com/resources/mensadb.
Collapse
Affiliation(s)
- Pedro Matos-Filipe
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3005-504, Portugal
| | - António J Preto
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3005-504, Portugal
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal
| | - Panagiotis I Koukos
- Bijvoet Centre for Biomolecular Research, Faculty of Science—Chemistry, Utrecht University, Utrecht, 3584, CH, Netherlands
| | - Joana Mourão
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3005-504, Portugal
| | - Alexandre M J J Bonvin
- Bijvoet Centre for Biomolecular Research, Faculty of Science—Chemistry, Utrecht University, Utrecht, 3584, CH, Netherlands
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Coimbra, 3000-456, Portugal
- Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
34
|
Pluska L, Jarosch E, Zauber H, Kniss A, Waltho A, Bagola K, von Delbrück M, Löhr F, Schulman BA, Selbach M, Dötsch V, Sommer T. The UBA domain of conjugating enzyme Ubc1/Ube2K facilitates assembly of K48/K63-branched ubiquitin chains. EMBO J 2021; 40:e106094. [PMID: 33576509 PMCID: PMC7957398 DOI: 10.15252/embj.2020106094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 12/23/2022] Open
Abstract
The assembly of a specific polymeric ubiquitin chain on a target protein is a key event in the regulation of numerous cellular processes. Yet, the mechanisms that govern the selective synthesis of particular polyubiquitin signals remain enigmatic. The homologous ubiquitin-conjugating (E2) enzymes Ubc1 (budding yeast) and Ube2K (mammals) exclusively generate polyubiquitin linked through lysine 48 (K48). Uniquely among E2 enzymes, Ubc1 and Ube2K harbor a ubiquitin-binding UBA domain with unknown function. We found that this UBA domain preferentially interacts with ubiquitin chains linked through lysine 63 (K63). Based on structural modeling, in vitro ubiquitination experiments, and NMR studies, we propose that the UBA domain aligns Ubc1 with K63-linked polyubiquitin and facilitates the selective assembly of K48/K63-branched ubiquitin conjugates. Genetic and proteomics experiments link the activity of the UBA domain, and hence the formation of this unusual ubiquitin chain topology, to the maintenance of cellular proteostasis.
Collapse
Affiliation(s)
- Lukas Pluska
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
| | - Ernst Jarosch
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
| | - Henrik Zauber
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
| | - Andreas Kniss
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic ResonanceGoethe UniversityFrankfurt am MainGermany
| | - Anita Waltho
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
| | - Katrin Bagola
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
| | | | - Frank Löhr
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic ResonanceGoethe UniversityFrankfurt am MainGermany
| | - Brenda A Schulman
- Department of Molecular Machines and SignalingMax Planck Institute of BiochemistryMartinsriedGermany
| | - Matthias Selbach
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
- Charité – Universitätsmedizin BerlinBerlinGermany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic ResonanceGoethe UniversityFrankfurt am MainGermany
| | - Thomas Sommer
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlin‐BuchGermany
- Institute for BiologyHumboldt‐Universität zu BerlinBerlinGermany
| |
Collapse
|
35
|
Sitani D, Giorgetti A, Alfonso-Prieto M, Carloni P. Robust principal component analysis-based prediction of protein-protein interaction hot spots. Proteins 2021; 89:639-647. [PMID: 33458895 DOI: 10.1002/prot.26047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 12/21/2022]
Abstract
Proteins often exert their function by binding to other cellular partners. The hot spots are key residues for protein-protein binding. Their identification may shed light on the impact of disease associated mutations on protein complexes and help design protein-protein interaction inhibitors for therapy. Unfortunately, current machine learning methods to predict hot spots, suffer from limitations caused by gross errors in the data matrices. Here, we present a novel data pre-processing pipeline that overcomes this problem by recovering a low rank matrix with reduced noise using Robust Principal Component Analysis. Application to existing databases shows the predictive power of the method.
Collapse
Affiliation(s)
- Divya Sitani
- JARA-Institute: Molecular Neuroscience and Neuroimaging, Institute for Neuroscience and Medicine INM-11/JARA-BRAIN Institute JBI-2, Forschungszentrum Jülich GmbH, Jülich, Germany.,Department of Biology, RWTH Aachen University, Aachen, Germany
| | - Alejandro Giorgetti
- Institute for Advanced Simulations IAS-5 / Institute for Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, Germany.,Department of Biotechnology, University of Verona, Verona, Italy
| | - Mercedes Alfonso-Prieto
- Institute for Advanced Simulations IAS-5 / Institute for Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, Germany.,Cécile and Oskar Vogt Institute for Brain Research, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Paolo Carloni
- JARA-Institute: Molecular Neuroscience and Neuroimaging, Institute for Neuroscience and Medicine INM-11/JARA-BRAIN Institute JBI-2, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute for Advanced Simulations IAS-5 / Institute for Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, Germany.,Department of Physics, RWTH Aachen University, Aachen, Germany.,JARA-HPC, IAS-5/INM-9 Computational Biomedicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| |
Collapse
|
36
|
SAAFEC-SEQ: A Sequence-Based Method for Predicting the Effect of Single Point Mutations on Protein Thermodynamic Stability. Int J Mol Sci 2021; 22:ijms22020606. [PMID: 33435356 PMCID: PMC7827184 DOI: 10.3390/ijms22020606] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 01/04/2023] Open
Abstract
Modeling the effect of mutations on protein thermodynamics stability is useful for protein engineering and understanding molecular mechanisms of disease-causing variants. Here, we report a new development of the SAAFEC method, the SAAFEC-SEQ, which is a gradient boosting decision tree machine learning method to predict the change of the folding free energy caused by amino acid substitutions. The method does not require the 3D structure of the corresponding protein, but only its sequence and, thus, can be applied on genome-scale investigations where structural information is very sparse. SAAFEC-SEQ uses physicochemical properties, sequence features, and evolutionary information features to make the predictions. It is shown to consistently outperform all existing state-of-the-art sequence-based methods in both the Pearson correlation coefficient and root-mean-squared-error parameters as benchmarked on several independent datasets. The SAAFEC-SEQ has been implemented into a web server and is available as stand-alone code that can be downloaded and embedded into other researchers’ code.
Collapse
|
37
|
Slater O, Miller B, Kontoyianni M. Decoding Protein-protein Interactions: An Overview. Curr Top Med Chem 2021; 20:855-882. [PMID: 32101126 DOI: 10.2174/1568026620666200226105312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022]
Abstract
Drug discovery has focused on the paradigm "one drug, one target" for a long time. However, small molecules can act at multiple macromolecular targets, which serves as the basis for drug repurposing. In an effort to expand the target space, and given advances in X-ray crystallography, protein-protein interactions have become an emerging focus area of drug discovery enterprises. Proteins interact with other biomolecules and it is this intricate network of interactions that determines the behavior of the system and its biological processes. In this review, we briefly discuss networks in disease, followed by computational methods for protein-protein complex prediction. Computational methodologies and techniques employed towards objectives such as protein-protein docking, protein-protein interactions, and interface predictions are described extensively. Docking aims at producing a complex between proteins, while interface predictions identify a subset of residues on one protein that could interact with a partner, and protein-protein interaction sites address whether two proteins interact. In addition, approaches to predict hot spots and binding sites are presented along with a representative example of our internal project on the chemokine CXC receptor 3 B-isoform and predictive modeling with IP10 and PF4.
Collapse
Affiliation(s)
- Olivia Slater
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Bethany Miller
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Maria Kontoyianni
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| |
Collapse
|
38
|
Preto AJ, Matos-Filipe P, de Almeida JG, Mourão J, Moreira IS. Predicting Hot Spots Using a Deep Neural Network Approach. Methods Mol Biol 2021; 2190:267-288. [PMID: 32804371 DOI: 10.1007/978-1-0716-0826-5_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Targeting protein-protein interactions is a challenge and crucial task of the drug discovery process. A good starting point for rational drug design is the identification of hot spots (HS) at protein-protein interfaces, typically conserved residues that contribute most significantly to the binding. In this chapter, we depict point-by-point an in-house pipeline used for HS prediction using only sequence-based features from the well-known SpotOn dataset of soluble proteins (Moreira et al., Sci Rep 7:8007, 2017), through the implementation of a deep neural network. The presented pipeline is divided into three steps: (1) feature extraction, (2) deep learning classification, and (3) model evaluation. We present all the available resources, including code snippets, the main dataset, and the free and open-source modules/packages necessary for full replication of the protocol. The users should be able to develop an HS prediction model with accuracy, precision, recall, and AUROC of 0.96, 0.93, 0.91, and 0.86, respectively.
Collapse
Affiliation(s)
- António J Preto
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Pedro Matos-Filipe
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José G de Almeida
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Joana Mourão
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Irina S Moreira
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- University of Coimbra, Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
39
|
Nandakumar R, Dinu V. Developing a machine learning model to identify protein–protein interaction hotspots to facilitate drug discovery. PeerJ 2020; 8:e10381. [PMID: 33354416 PMCID: PMC7727375 DOI: 10.7717/peerj.10381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/27/2020] [Indexed: 02/01/2023] Open
Abstract
Throughout the history of drug discovery, an enzymatic-based approach for identifying new drug molecules has been primarily utilized. Recently, protein–protein interfaces that can be disrupted to identify small molecules that could be viable targets for certain diseases, such as cancer and the human immunodeficiency virus, have been identified. Existing studies computationally identify hotspots on these interfaces, with most models attaining accuracies of ~70%. Many studies do not effectively integrate information relating to amino acid chains and other structural information relating to the complex. Herein, (1) a machine learning model has been created and (2) its ability to integrate multiple features, such as those associated with amino-acid chains, has been evaluated to enhance the ability to predict protein–protein interface hotspots. Virtual drug screening analysis of a set of hotspots determined on the EphB2-ephrinB2 complex has also been performed. The predictive capabilities of this model offer an AUROC of 0.842, sensitivity/recall of 0.833, and specificity of 0.850. Virtual screening of a set of hotspots identified by the machine learning model developed in this study has identified potential medications to treat diseases caused by the overexpression of the EphB2-ephrinB2 complex, including prostate, gastric, colorectal and melanoma cancers which are linked to EphB2 mutations. The efficacy of this model has been demonstrated through its successful ability to predict drug-disease associations previously identified in literature, including cimetidine, idarubicin, pralatrexate for these conditions. In addition, nadolol, a beta blocker, has also been identified in this study to bind to the EphB2-ephrinB2 complex, and the possibility of this drug treating multiple cancers is still relatively unexplored.
Collapse
|
40
|
Buckton LK, Rahimi MN, McAlpine SR. Cyclic Peptides as Drugs for Intracellular Targets: The Next Frontier in Peptide Therapeutic Development. Chemistry 2020; 27:1487-1513. [PMID: 32875673 DOI: 10.1002/chem.201905385] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 08/26/2020] [Indexed: 12/18/2022]
Abstract
Developing macrocyclic peptides that can reach intracellular targets is a significant challenge. This review discusses the most recent strategies used to develop cell permeable cyclic peptides that maintain binding to their biological target inside the cell. Macrocyclic peptides are unique from small molecules because traditional calculated physical properties are unsuccessful for predicting cell membrane permeability. Peptide synthesis and experimental membrane permeability is the only strategy that effectively differentiates between cell permeable and cell impermeable molecules. Discussed are chemical strategies, including backbone N-methylation and stereochemical changes, which have produced molecular scaffolds with improved cell permeability. However, these improvements often come at the expense of biological activity as chemical modifications alter the peptide conformation, frequently impacting the compound's ability to bind to the target. Highlighted is the most promising approach, which involves side-chain alterations that improve cell permeability without impact binding events.
Collapse
Affiliation(s)
- Laura K Buckton
- Department of Chemistry, University of New South Wales, Sydney, Gate 2 High Street, SEB 701, Kensington, NSW, 2052, Australia
| | - Marwa N Rahimi
- Department of Chemistry, University of New South Wales, Sydney, Gate 2 High Street, SEB 701, Kensington, NSW, 2052, Australia
| | - Shelli R McAlpine
- Department of Chemistry, University of New South Wales, Sydney, Gate 2 High Street, SEB 701, Kensington, NSW, 2052, Australia
| |
Collapse
|
41
|
Syahbanu F, Giriwono PE, Tjandrawinata RR, Suhartono MT. Molecular analysis of a fibrin-degrading enzyme from Bacillus subtilis K2 isolated from the Indonesian soybean-based fermented food moromi. Mol Biol Rep 2020; 47:8553-8563. [PMID: 33111172 DOI: 10.1007/s11033-020-05898-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/03/2020] [Indexed: 10/23/2022]
Abstract
The screening of proteolytic and fibrinolytic bacteria from moromi (an Indonesian soybean-based fermented food) yielded a number of isolates. Based on morphological and biochemical analyses and sequencing of the 16S rRNA gene, the isolate that exhibited the highest proteolytic and fibrinolytic activity was identified as Bacillus subtilis K2. The study was performed to analyze molecular characteristic of a fibrin-degrading enzyme from B. subtilis K2. BLASTn analysis of the nucleotide sequence encoding this fibrinolytic protein demonstrated 73.6% homology with the gene encoding the fibrin-degrading enzyme nattokinase of the B. subtilis subsp. natto, which was isolated from fermented soybean in Japan. An analysis of the putative amino-acid sequence of this protein indicated that it is a serine protease enzyme with aspartate, histidine, and serine in the catalytic triad. This enzyme was determined to be a 26-kDa molecule, as confirmed with a zymogram assay. Further bioinformatic analysis using Protparam demonstrated that the enzyme has a pI of 6.02, low instability index, high aliphatic index, and low GRAVY value. Molecular docking analysis using HADDOCK indicated that there are favorable interactions between subtilisin K2 and the fibrin substrate, as demonstrated by a high binding affinity (ΔG: - 19.4 kcal/mol) and low Kd value (6.3E-15 M). Overall, the study concluded that subtilisin K2 belong to serine protease enzyme has strong interactions with its fibrin substrate and fibrin can be rapidly degraded by this enzyme, suggesting its application as a treatment for thrombus diseases.
Collapse
Affiliation(s)
- Fathma Syahbanu
- Department of Food Science and Technology, IPB University (Bogor Agricultural University), Dramaga, P.O. BOX 220, Bogor, Indonesia
| | - Puspo Edi Giriwono
- Department of Food Science and Technology, IPB University (Bogor Agricultural University), Dramaga, P.O. BOX 220, Bogor, Indonesia
| | | | - Maggy T Suhartono
- Department of Food Science and Technology, IPB University (Bogor Agricultural University), Dramaga, P.O. BOX 220, Bogor, Indonesia.
| |
Collapse
|
42
|
Bouricha EM, Hakmi M, Akachar J, Belyamani L, Ibrahimi A. In silico analysis of ACE2 orthologues to predict animal host range with high susceptibility to SARS-CoV-2. 3 Biotech 2020; 10:483. [PMID: 33101829 PMCID: PMC7577366 DOI: 10.1007/s13205-020-02471-3] [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: 06/22/2020] [Accepted: 10/07/2020] [Indexed: 11/01/2022] Open
Abstract
SARS-CoV-2, which causes severe pneumonia epidemics, probably originated from Chinese horseshoe bats, but the intermediate and host range is still unknown. ACE2 is the entry receptor for SARS-CoV-2. The binding capacity of SARS-CoV-2 spike protein to ACE2 is the critical determinant of viral host range and cross-species infection. Here, we used an in silico approach to predict the potential animals range with high susceptibility to SARS-CoV-2 by modelling and studying the Spike-ACE2 interaction of 22 domestic and wild animals. Our results showed that all studied animals are potentially susceptible to SARS-CoV-2 infection with a slight difference in the binding affinity and stability of their ACE2-RBD complexes. Furthermore, we identified a specific substitution of tyrosine to histidine at position 41 in ACE2 that likely reduces the affinity to SARS-CoV-2 in horses and greater horseshoe bats. These results may help to provide important insights into SARS-CoV-2 host range which will make it possible to control the spread of the virus and identify animal models that could be used for screening antiviral drugs or vaccine candidates against SARS-CoV-2.
Collapse
Affiliation(s)
- El Mehdi Bouricha
- Medical Biotechnology Laboratory (MedBiotech), Rabat Medical and Pharmacy School, Mohammed Vth University in Rabat, Rabat, Morocco
| | - Mohammed Hakmi
- Medical Biotechnology Laboratory (MedBiotech), Rabat Medical and Pharmacy School, Mohammed Vth University in Rabat, Rabat, Morocco
| | - Jihane Akachar
- Medical Biotechnology Laboratory (MedBiotech), Rabat Medical and Pharmacy School, Mohammed Vth University in Rabat, Rabat, Morocco
| | - Lahcen Belyamani
- Emergency Department, Military Hospital Mohammed V, Rabat Medical and Pharmacy School, Mohammed Vth University in Rabat, Rabat, Morocco
| | - Azeddine Ibrahimi
- Medical Biotechnology Laboratory (MedBiotech), Rabat Medical and Pharmacy School, Mohammed Vth University in Rabat, Rabat, Morocco
| |
Collapse
|
43
|
Preto AJ, Moreira IS. SPOTONE: Hot Spots on Protein Complexes with Extremely Randomized Trees via Sequence-Only Features. Int J Mol Sci 2020; 21:ijms21197281. [PMID: 33019775 PMCID: PMC7582262 DOI: 10.3390/ijms21197281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 01/02/2023] Open
Abstract
Protein Hot-Spots (HS) are experimentally determined amino acids, key to small ligand binding and tend to be structural landmarks on protein–protein interactions. As such, they were extensively approached by structure-based Machine Learning (ML) prediction methods. However, the availability of a much larger array of protein sequences in comparison to determined tree-dimensional structures indicates that a sequence-based HS predictor has the potential to be more useful for the scientific community. Herein, we present SPOTONE, a new ML predictor able to accurately classify protein HS via sequence-only features. This algorithm shows accuracy, AUROC, precision, recall and F1-score of 0.82, 0.83, 0.91, 0.82 and 0.85, respectively, on an independent testing set. The algorithm is deployed within a free-to-use webserver, only requiring the user to submit a FASTA file with one or more protein sequences.
Collapse
Affiliation(s)
- A. J. Preto
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
| | - Irina S. Moreira
- Department of Life Sciences, Center for Neuroscience and Cell Biology, Coimbra University, 3000-456 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
44
|
Koirala M, Alexov E. Ab-initio binding of barnase–barstar with DelPhiForce steered Molecular Dynamics (DFMD) approach. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2020. [DOI: 10.1142/s0219633620500169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Receptor–ligand interactions are involved in various biological processes, therefore understanding the binding mechanism and ability to predict the binding mode are essential for many biological investigations. While many computational methods exist to predict the 3D structure of the corresponding complex provided the knowledge of the monomers, here we use the newly developed DelPhiForce steered Molecular Dynamics (DFMD) approach to model the binding of barstar to barnase to demonstrate that first-principles methods are also capable of modeling the binding. Essential component of DFMD approach is enhancing the role of long-range electrostatic interactions to provide guiding force of the monomers toward their correct binding orientation and position. Thus, it is demonstrated that the DFMD can successfully dock barstar to barnase even if the initial positions and orientations of both are completely different from the correct ones. Thus, the electrostatics provides orientational guidance along with pulling force to deliver the ligand in close proximity to the receptor.
Collapse
Affiliation(s)
- Mahesh Koirala
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
45
|
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
|
46
|
da Silva FCV, Pessoa Costa E, Moreira Gomes V, de Oliveira Carvalho A. Inhibition mechanism of human salivary α-amylase by lipid transfer protein from Vigna unguiculata. Comput Biol Chem 2020; 85:107193. [DOI: 10.1016/j.compbiolchem.2019.107193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 01/09/2023]
|
47
|
Barreto CAV, Baptista SJ, Preto AJ, Matos-Filipe P, Mourão J, Melo R, Moreira I. Prediction and targeting of GPCR oligomer interfaces. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 169:105-149. [PMID: 31952684 DOI: 10.1016/bs.pmbts.2019.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
GPCR oligomerization has emerged as a hot topic in the GPCR field in the last years. Receptors that are part of these oligomers can influence each other's function, although it is not yet entirely understood how these interactions work. The existence of such a highly complex network of interactions between GPCRs generates the possibility of alternative targets for new therapeutic approaches. However, challenges still exist in the characterization of these complexes, especially at the interface level. Different experimental approaches, such as FRET or BRET, are usually combined to study GPCR oligomer interactions. Computational methods have been applied as a useful tool for retrieving information from GPCR sequences and the few X-ray-resolved oligomeric structures that are accessible, as well as for predicting new and trustworthy GPCR oligomeric interfaces. Machine-learning (ML) approaches have recently helped with some hindrances of other methods. By joining and evaluating multiple structure-, sequence- and co-evolution-based features on the same algorithm, it is possible to dilute the issues of particular structures and residues that arise from the experimental methodology into all-encompassing algorithms capable of accurately predict GPCR-GPCR interfaces. All these methods used as a single or a combined approach provide useful information about GPCR oligomerization and its role in GPCR function and dynamics. Altogether, we present experimental, computational and machine-learning methods used to study oligomers interfaces, as well as strategies that have been used to target these dynamic complexes.
Collapse
Affiliation(s)
- Carlos A V Barreto
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Salete J Baptista
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, LRS, Portugal
| | - António José Preto
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Pedro Matos-Filipe
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Joana Mourão
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rita Melo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, LRS, Portugal
| | - Irina Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Science and Technology Faculty, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
48
|
Díaz-Valle A, Falcón-González JM, Carrillo-Tripp M. Hot Spots and Their Contribution to the Self-Assembly of the Viral Capsid: In Silico Prediction and Analysis. Int J Mol Sci 2019; 20:E5966. [PMID: 31783519 PMCID: PMC6928768 DOI: 10.3390/ijms20235966] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
The viral capsid is a macromolecular complex formed by a defined number of self-assembled proteins, which, in many cases, are biopolymers with an identical amino acid sequence. Specific protein-protein interactions (PPI) drive the capsid self-assembly process, leading to several distinct protein interfaces. Following the PPI hot spot hypothesis, we present a conservation-based methodology to identify those interface residues hypothesized to be crucial elements on the self-assembly and thermodynamic stability of the capsid. We validate the predictions through a rigorous physical framework which integrates molecular dynamics simulations and free energy calculations by Umbrella sampling and the potential of mean force using an all-atom molecular representation of the capsid proteins of an icosahedral virus in an explicit solvent. Our results show that a single mutation in any of the structure-conserved hot spots significantly perturbs the quaternary protein-protein interaction, decreasing the absolute value of the binding free energy, without altering the protein's secondary nor tertiary structure. Our conservation-based hot spot prediction methodology can lead to strategies to rationally modulate the capsid's thermodynamic properties.
Collapse
Affiliation(s)
- Armando Díaz-Valle
- Biomolecular Diversity Laboratory, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional Unidad Monterrey, Vía del Conocimiento 201, Parque PIIT, C.P. 66600 Apodaca, Nuevo León, Mexico;
| | - José Marcos Falcón-González
- Unidad Profesional Interdisciplinaria de Ingeniería Campus Guanajuato, Instituto Politécnico Nacional, Av. Mineral de Valenciana No. 200, Col. Fraccionamiento Industrial Puerto Interior, C.P. 36275 Silao de la Victoria, Guanajuato, Mexico;
| | - Mauricio Carrillo-Tripp
- Biomolecular Diversity Laboratory, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional Unidad Monterrey, Vía del Conocimiento 201, Parque PIIT, C.P. 66600 Apodaca, Nuevo León, Mexico;
| |
Collapse
|
49
|
Bonetta R, Valentino G. Machine learning techniques for protein function prediction. Proteins 2019; 88:397-413. [PMID: 31603244 DOI: 10.1002/prot.25832] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/05/2019] [Accepted: 09/17/2019] [Indexed: 12/17/2022]
Abstract
Proteins play important roles in living organisms, and their function is directly linked with their structure. Due to the growing gap between the number of proteins being discovered and their functional characterization (in particular as a result of experimental limitations), reliable prediction of protein function through computational means has become crucial. This paper reviews the machine learning techniques used in the literature, following their evolution from simple algorithms such as logistic regression to more advanced methods like support vector machines and modern deep neural networks. Hyperparameter optimization methods adopted to boost prediction performance are presented. In parallel, the metamorphosis in the features used by these algorithms from classical physicochemical properties and amino acid composition, up to text-derived features from biomedical literature and learned feature representations using autoencoders, together with feature selection and dimensionality reduction techniques, are also reviewed. The success stories in the application of these techniques to both general and specific protein function prediction are discussed.
Collapse
Affiliation(s)
- Rosalin Bonetta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Gianluca Valentino
- Department of Communications and Computer Engineering, University of Malta, Msida, Malta
| |
Collapse
|
50
|
Yang W, Sun X, Zhang C, Lai L. Discovery of novel helix binding sites at protein-protein interfaces. Comput Struct Biotechnol J 2019; 17:1396-1403. [PMID: 31768230 PMCID: PMC6872852 DOI: 10.1016/j.csbj.2019.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 01/09/2023] Open
Abstract
Protein-protein interactions (PPIs) play a key role in numerous biological processes. Many efforts have been undertaken to develop PPI modulators for therapeutic applications; however, to date, most of the peptide binders designed to target PPIs are derived from native binding helices or using the native helix binding site, which has limited the applications of protein-protein interface binding peptide design. Here, we developed a general computational algorithm, HPer (Helix Positioner), that locates single-helix binding sites at protein-protein interfaces based on the structure of protein targets. HPer performed well on known single-helix-mediated PPIs and recaptured the key interactions and hot-spot residues of native helical binders. We also screened non-helical-mediated PPIs in the PDBbind database and identified 17 PPIs that were suitable for helical peptide binding, and the helical binding sites in these PPIs were also predicted for designing novel peptide ligands. The L2 domain of EGFR, which was the top ranked, was selected as an example to show the protocol and results of designing novel helical peptide ligands on the searched binding site. The binding stability of the designed sequences were further investigated using molecular dynamics simulations.
Collapse
Affiliation(s)
- Wei Yang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Center for Quantitative Biology, AAIS, Peking University, Beijing 100871, China
| | - Xiangyu Sun
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Changsheng Zhang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Luhua Lai
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, AAIS, Peking University, Beijing 100084, China
- Center for Quantitative Biology, AAIS, Peking University, Beijing 100871, China
| |
Collapse
|