1
|
Li Q, Zhao Y, Chordia MD, Xia X, Zhang B, Zheng H. Enhanced prediction of antigen and antibody binding interface using ESM-2 and Bi-LSTM. Hum Immunol 2025; 86:111304. [PMID: 40188508 DOI: 10.1016/j.humimm.2025.111304] [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: 10/28/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/08/2025]
Abstract
The binding interface between antigens and antibodies is pivotal in humoral immune responses and provides crucial effective defense against pathogens and exogenous threats. Existing predictive computational methodologies, including structure-based and sequence-based approaches, offer valuable insights but face challenges such as unknown antigen structures and reliance on manually curated features. Most current methods primarily predict antigen epitope, often neglecting the specific molecular epitope-paratope interactions essential for immune efficacy. In this study, we introduce a novel approach EPP (Epitope-Paratope Predictor), using the ESM-2 protein language model as a feature encoder and a Bi-LSTM network to predict epitope-paratope interactions. Our method processes antigen and antibody sequences as inputs, leveraging a novel dataset strategy and encoding protein representations to enhance prediction accuracy. The results demonstrate a significant improvement in prediction accuracy compared to existing methods, highlighting the importance of protein feature encoder and temporal dependencies within sequences. The model's performance in different antigen clusters is analyzed, while those predictions are compared with that from AlphaFold3 and Dock method. Our method validation shows superior performance in recognizing distinctive epitopes of the same antigen when bound to different antibodies. This approach offers a new strategy for an in-depth understanding of antigen-antibody interactions, essential for an array of pioneer projects, such as structure-guided design and affinity maturation for precision antibodies targeting a given epitope.
Collapse
Affiliation(s)
- Qianying Li
- Hunan University College of Biology, Changsha, Hunan 410082, China
| | - Yanmin Zhao
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China; Shenzhen Tributary Biologics LLC, Shenzhen, Guangdong 518000, China
| | - Mahendra D Chordia
- Department of Chemistry, University of Virginia, Charlottesville, VA 22908, USA
| | - Xiuming Xia
- Department of Computer Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Bo Zhang
- College of Computing and Data Science, Nanyang Technological University, Singapore
| | - Heping Zheng
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China; Shenzhen Tributary Biologics LLC, Shenzhen, Guangdong 518000, China.
| |
Collapse
|
2
|
Chen Z, Xin C, Zhang S, Zhan Q, Zhang F, Liu Y, Yang X, Yang X, Ren Z. Unraveling the immunogenic landscape: A quest for B-cell linear epitopes in the African swine fever virus H339R protein. Int J Biol Macromol 2025; 310:142944. [PMID: 40210029 DOI: 10.1016/j.ijbiomac.2025.142944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
African swine fever (ASF) is a highly contagious viral disease causing severe economic losses in the global swine industry. The development of effective diagnostic tools and vaccines is crucial for the control and prevention of ASF. In this study, we aimed to identify and characterize linear B-cell epitopes within the African swine fever virus (ASFV) H339R protein, a potential diagnostic and vaccine target. Using monoclonal antibodies (mAbs) and enzyme-linked immunosorbent assay (ELISA)-based mapping, we identified four distinct linear B-cell epitopes in the H339R protein. These epitopes exhibited specific reactivity with ASFV-positive swine sera in western blot (WB) and indirect ELISA assays. Alanine-scanning mutagenesis revealed critical amino acid residues within each epitope that contribute to antibody recognition. The identification and characterization of these linear B-cell epitopes in the ASFV H339R protein provide valuable insights into the antigenic structure of the virus and lay the foundation for the development of novel diagnostic tools and epitope-based vaccines for ASF. These findings contribute to control and prevent this economically significant disease and to safeguard the global swine industry.
Collapse
Affiliation(s)
- Zhuting Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Cheng Xin
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China
| | - Shuang Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qinyi Zhan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Zhouzheng Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
3
|
Dewaker V, Morya VK, Kim YH, Park ST, Kim HS, Koh YH. Revolutionizing oncology: the role of Artificial Intelligence (AI) as an antibody design, and optimization tools. Biomark Res 2025; 13:52. [PMID: 40155973 PMCID: PMC11954232 DOI: 10.1186/s40364-025-00764-4] [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: 02/25/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
Antibodies play a crucial role in defending the human body against diseases, including life-threatening conditions like cancer. They mediate immune responses against foreign antigens and, in some cases, self-antigens. Over time, antibody-based technologies have evolved from monoclonal antibodies (mAbs) to chimeric antigen receptor T cells (CAR-T cells), significantly impacting biotechnology, diagnostics, and therapeutics. Although these advancements have enhanced therapeutic interventions, the integration of artificial intelligence (AI) is revolutionizing antibody design and optimization. This review explores recent AI advancements, including large language models (LLMs), diffusion models, and generative AI-based applications, which have transformed antibody discovery by accelerating de novo generation, enhancing immune response precision, and optimizing therapeutic efficacy. Through advanced data analysis, AI enables the prediction and design of antibody sequences, 3D structures, complementarity-determining regions (CDRs), paratopes, epitopes, and antigen-antibody interactions. These AI-powered innovations address longstanding challenges in antibody development, significantly improving speed, specificity, and accuracy in therapeutic design. By integrating computational advancements with biomedical applications, AI is driving next-generation cancer therapies, transforming precision medicine, and enhancing patient outcomes.
Collapse
Affiliation(s)
- Varun Dewaker
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
| | - Vivek Kumar Morya
- Department of Orthopedic Surgery, Hallym University Dongtan Sacred Hospital, Hwaseong-Si, 18450, Republic of Korea
| | - Yoo Hee Kim
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea.
- Department of Internal Medicine, Division of Hemato-Oncology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea.
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea.
| | - Young Ho Koh
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea.
| |
Collapse
|
4
|
Li Y, Zhao G, Zhang Y, Xia L, Cheng Y, Ma J, Wang H, Yan Y, Wang Z, Sun J. Bacteriophage M13KE as a nanoparticle platform to display and deliver a pathogenic epitope: Development of an effective porcine epidemic diarrhoea virus vaccine. Microb Pathog 2025; 200:107325. [PMID: 39864763 DOI: 10.1016/j.micpath.2025.107325] [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: 07/12/2024] [Revised: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Porcine epidemic diarrhoea virus (PEDV) is a porcine enteric coronavirus, outbreaks and epidemics of which have caused huge economic losses to the livestock industry. The disadvantage of existing PEDV vaccines is that the unstable efficacy and high cost limit their widespread use. Therefore, there is an urgent need to develop a recombinant transgenic vaccine candidate for PEDV. In this study, three linear epitopes on the PEDV spike (S) were screened using peptide scanning. The screened epitopes were linked to targeting peptides for lung and intestinal epithelial cells, respectively, and displayed on the M13KE phage to form recombinant phage nanoparticles. Active immunisation experiments showed that a single B-cell epitope delivered by M13KE phage nanoparticles induced the production of specific neutralising antibodies against PEDV in mice. After PEDV stimulation, the immunised mice had significantly higher levels of interferon-γ (IFN-γ) than the control group. Simultaneously, PEDV stimulation caused lymphocyte activation and proliferation in the immunised mice, which is a typical immune response to viral infections. These results suggest that a single linear antigenic epitope delivered by M13KE phage nanoparticles induces significant humoral and cellular immune responses. The constructed recombinant phage nanoparticles are expected to be potential vaccine candidates for PEDV.
Collapse
MESH Headings
- Animals
- Porcine epidemic diarrhea virus/immunology
- Porcine epidemic diarrhea virus/genetics
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Coronavirus Infections/prevention & control
- Coronavirus Infections/veterinary
- Coronavirus Infections/immunology
- Nanoparticles
- Antibodies, Viral/blood
- Mice
- Antibodies, Neutralizing/blood
- Swine
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Interferon-gamma/metabolism
- Mice, Inbred BALB C
- Swine Diseases/prevention & control
- Swine Diseases/virology
- Swine Diseases/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/administration & dosage
- Epitopes/immunology
- Epitopes/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/genetics
- Female
Collapse
Affiliation(s)
- Yan Li
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Guoqing Zhao
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Yumin Zhang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Lu Xia
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Yuqiang Cheng
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Jingjiao Ma
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Henan Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Yaxian Yan
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Zhaofei Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China.
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 201100, China.
| |
Collapse
|
5
|
Hamza S, Shakirova V, Khaertynova I, Markelova M, Saxena PV, Sharma D, Kaushal N, Gupta Y, Garanina E, Pavelkina V, Khaiboullina S, Martynova E, Rizvanov A, Baranwal M. Identification and validation of cross-reactivity of anti-Thailand orthohantavirus nucleocapsid peptides. Hum Immunol 2024; 85:111157. [PMID: 39423729 DOI: 10.1016/j.humimm.2024.111157] [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: 06/24/2024] [Revised: 08/24/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
A Thailand orthohantavirus (THAIV) is endemic in Southeast Asia. This assumption is supported by isolation of THAIV from local small mammals. Also, anti-orthohantavirus antibodies were detected in human serum. However, our understanding of THAIV cross-reactivity with antibodies against other orthohantaviruses remains largely unknown. We used the in-silico approach to identify the cross-reactive immunogenic peptides of THAIV. The immunogenicity of these peptides was tested using convalescent serum from patients infected with Puumala (PUUV), Hantaan (HNTV) and Dobrava (DOBV) orthohantaviruses. We identified three THAIV peptides reacting with orthohantavirus convalescent serum. P1 peptide was reactive with serum from patients infected with PUUV, HNTV and DOBV. These peptides were found to be non-allergenic. Molecular docking and population coverage analysis revealed the potential of selected peptides to interact with diverse HLA alleles worldwide. Our data indicate that THAIV peptides could be used to develop diagnostics for orthohantaviruses circulating in Southeast Asia.
Collapse
Affiliation(s)
| | - Venara Shakirova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | - Ilsiyar Khaertynova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | | | - Prakhar Vaidant Saxena
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Diksha Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Neha Kaushal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Yogita Gupta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | | | - Vera Pavelkina
- Infectious Diseases Department, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | | | | | | | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India.
| |
Collapse
|
6
|
Kumar N, Tripathi S, Sharma N, Patiyal S, Devi NL, Raghava GPS. A method for predicting linear and conformational B-cell epitopes in an antigen from its primary sequence. Comput Biol Med 2024; 170:108083. [PMID: 38295479 DOI: 10.1016/j.compbiomed.2024.108083] [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: 06/21/2023] [Revised: 12/26/2023] [Accepted: 01/27/2024] [Indexed: 02/02/2024]
Abstract
B-cell is an essential component of the immune system that plays a vital role in providing the immune response against any pathogenic infection by producing antibodies. Existing methods either predict linear or conformational B-cell epitopes in an antigen. In this study, a single method was developed for predicting both types (linear/conformational) of B-cell epitopes. The dataset used in this study contains 3875 B-cell epitopes and 3996 non-B-cell epitopes, where B-cell epitopes consist of both linear and conformational B-cell epitopes. Our primary analysis indicates that certain residues (like Asp, Glu, Lys, and Asn) are more prominent in B-cell epitopes. We developed machine-learning based methods using different types of sequence composition and achieved the highest AUROC of 0.80 using dipeptide composition. In addition, models were developed on selected features, but no further improvement was observed. Our similarity-based method implemented using BLAST shows a high probability of correct prediction with poor sensitivity. Finally, we developed a hybrid model that combines alignment-free (dipeptide based random forest model) and alignment-based (BLAST-based similarity) models. Our hybrid model attained a maximum AUROC of 0.83 with an MCC of 0.49 on the independent dataset. Our hybrid model performs better than existing methods on an independent dataset used in this study. All models were trained and tested on 80 % of the data using a cross-validation technique, and the final model was evaluated on 20 % of the data, called an independent or validation dataset. A webserver and standalone package named "CLBTope" has been developed for predicting, designing, and scanning B-cell epitopes in an antigen sequence available at (https://webs.iiitd.edu.in/raghava/clbtope/).
Collapse
Affiliation(s)
- Nishant Kumar
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi, 110020, India.
| | - Sadhana Tripathi
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi, 110020, India.
| | - Neelam Sharma
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi, 110020, India.
| | - Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi, 110020, India.
| | - Naorem Leimarembi Devi
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi, 110020, India.
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi, 110020, India.
| |
Collapse
|
7
|
Israeli S, Louzoun Y. Single-residue linear and conformational B cell epitopes prediction using random and ESM-2 based projections. Brief Bioinform 2024; 25:bbae084. [PMID: 38487845 PMCID: PMC10940830 DOI: 10.1093/bib/bbae084] [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: 11/06/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 03/18/2024] Open
Abstract
B cell epitope prediction methods are separated into linear sequence-based predictors and conformational epitope predictions that typically use the measured or predicted protein structure. Most linear predictions rely on the translation of the sequence to biologically based representations and the applications of machine learning on these representations. We here present CALIBER 'Conformational And LInear B cell Epitopes pRediction', and show that a bidirectional long short-term memory with random projection produces a more accurate prediction (test set AUC=0.789) than all current linear methods. The same predictor when combined with an Evolutionary Scale Modeling-2 projection also improves on the state of the art in conformational epitopes (AUC = 0.776). The inclusion of the graph of the 3D distances between residues did not increase the prediction accuracy. However, the long-range sequence information was essential for high accuracy. While the same model structure was applicable for linear and conformational epitopes, separate training was required for each. Combining the two slightly increased the linear accuracy (AUC 0.775 versus 0.768) and reduced the conformational accuracy (AUC = 0.769).
Collapse
Affiliation(s)
- Sapir Israeli
- Department of Mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Yoram Louzoun
- Department of Mathematics, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
8
|
Ramprasadh SV, Rajakumar S, Srinivasan S, Susha D, Sharma S, Chourasiya R. Computer-Aided Multi-Epitope Based Vaccine Design Against Monkeypox Virus Surface Protein A30L: An Immunoinformatics Approach. Protein J 2023; 42:645-663. [PMID: 37615828 DOI: 10.1007/s10930-023-10150-4] [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] [Accepted: 08/05/2023] [Indexed: 08/25/2023]
Abstract
Monkeypox, a viral zoonotic disease resembling smallpox, has emerged as a significant national epidemic primarily in Africa. Nevertheless, the recent global dissemination of this pathogen has engendered apprehension regarding its capacity to metamorphose into a sweeping pandemic. To effectively combat this menace, a multi-epitope vaccine has been meticulously engineered with the specific aim of targeting the cell envelope protein of Monkeypox virus (MPXV), thereby stimulating a potent immunological response while mitigating untoward effects. This new vaccine uses T-cell and B-cell epitopes from a highly antigenic, non-allergenic, non-toxic, conserved, and non-homologous A30L protein to provide protection against the virus. In order to ascertain the vaccine design with the utmost efficacy, protein-protein docking methodologies were employed to anticipate the intricate interactions with Toll-like receptors (TLR) 2, 3, 4, 6, and 8. This meticulous approach led the researchers to discern an optimal vaccine architecture, bolstered by affirmative prognostications derived from both molecular dynamics (MD) simulations and immune simulations. The current research findings indicate that the peptides ATHAAFEYSK, FFIVVATAAV, and MNSLSIFFV exhibited antigenic properties and were determined to be non-allergenic and non-toxic. Through the utilization of codon optimization and in-silico cloning techniques, our investigation revealed that the prospective vaccine exhibited a remarkable expression level within Escherichia coli. Moreover, upon conducting immune simulations, we observed the induction of a robust immune response characterized by elevated levels of both B-cell and T-cell mediated immunity. Moreover, as the initial prediction with in-silico techniques has yielded promising results these epitope-based vaccines can be recommended to in vitro and in silico studies to validate their immunogenic properties.
Collapse
Affiliation(s)
- S V Ramprasadh
- Department of Bioinformatics, BioNome, Bangalore, 560043, India
| | | | - S Srinivasan
- Department of Bioinformatics, BioNome, Bangalore, 560043, India
| | - D Susha
- Department of Bioinformatics, BioNome, Bangalore, 560043, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bangalore, 560043, India.
| | | |
Collapse
|
9
|
Kumar N, Bajiya N, Patiyal S, Raghava GPS. Multi-perspectives and challenges in identifying B-cell epitopes. Protein Sci 2023; 32:e4785. [PMID: 37733481 PMCID: PMC10578127 DOI: 10.1002/pro.4785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023]
Abstract
The identification of B-cell epitopes (BCEs) in antigens is a crucial step in developing recombinant vaccines or immunotherapies for various diseases. Over the past four decades, numerous in silico methods have been developed for predicting BCEs. However, existing reviews have only covered specific aspects, such as the progress in predicting conformational or linear BCEs. Therefore, in this paper, we have undertaken a systematic approach to provide a comprehensive review covering all aspects associated with the identification of BCEs. First, we have covered the experimental techniques developed over the years for identifying linear and conformational epitopes, including the limitations and challenges associated with these techniques. Second, we have briefly described the historical perspectives and resources that maintain experimentally validated information on BCEs. Third, we have extensively reviewed the computational methods developed for predicting conformational BCEs from the structure of the antigen, as well as the methods for predicting conformational epitopes from the sequence. Fourth, we have systematically reviewed the in silico methods developed in the last four decades for predicting linear or continuous BCEs. Finally, we have discussed the overall challenge of identifying continuous or conformational BCEs. In this review, we only listed major computational resources; a complete list with the URL is available from the BCinfo website (https://webs.iiitd.edu.in/raghava/bcinfo/).
Collapse
Affiliation(s)
- Nishant Kumar
- Department of Computational BiologyIndraprastha Institute of Information TechnologyNew DelhiIndia
| | - Nisha Bajiya
- Department of Computational BiologyIndraprastha Institute of Information TechnologyNew DelhiIndia
| | - Sumeet Patiyal
- Department of Computational BiologyIndraprastha Institute of Information TechnologyNew DelhiIndia
| | - Gajendra P. S. Raghava
- Department of Computational BiologyIndraprastha Institute of Information TechnologyNew DelhiIndia
| |
Collapse
|
10
|
Desta IT, Kotelnikov S, Jones G, Ghani U, Abyzov M, Kholodov Y, Standley DM, Beglov D, Vajda S, Kozakov D. The ClusPro AbEMap web server for the prediction of antibody epitopes. Nat Protoc 2023; 18:1814-1840. [PMID: 37188806 PMCID: PMC10898366 DOI: 10.1038/s41596-023-00826-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/19/2023] [Indexed: 05/17/2023]
Abstract
Antibodies play an important role in the immune system by binding to molecules called antigens at their respective epitopes. These interfaces or epitopes are structural entities determined by the interactions between an antibody and an antigen, making them ideal systems to analyze by using docking programs. Since the advent of high-throughput antibody sequencing, the ability to perform epitope mapping using only the sequence of the antibody has become a high priority. ClusPro, a leading protein-protein docking server, together with its template-based modeling version, ClusPro-TBM, have been re-purposed to map epitopes for specific antibody-antigen interactions by using the Antibody Epitope Mapping server (AbEMap). ClusPro-AbEMap offers three different modes for users depending on the information available on the antibody as follows: (i) X-ray structure, (ii) computational/predicted model of the structure or (iii) only the amino acid sequence. The AbEMap server presents a likelihood score for each antigen residue of being part of the epitope. We provide detailed information on the server's capabilities for the three options and discuss how to obtain the best results. In light of the recent introduction of AlphaFold2 (AF2), we also show how one of the modes allows users to use their AF2-generated antibody models as input. The protocol describes the relative advantages of the server compared to other epitope-mapping tools, its limitations and potential areas of improvement. The server may take 45-90 min depending on the size of the proteins.
Collapse
Affiliation(s)
- Israel T Desta
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sergei Kotelnikov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - George Jones
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Usman Ghani
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | | | | | - Daron M Standley
- Department of Genome Informatics, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Dmitri Beglov
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
11
|
Rashid S, Ng TA, Kwoh CK. Jupytope: computational extraction of structural properties of viral epitopes. Brief Bioinform 2022; 23:6696137. [PMID: 36094101 DOI: 10.1093/bib/bbac362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022] Open
Abstract
Epitope residues located on viral surface proteins are of immense interest in immunology and related applications such as vaccine development, disease diagnosis and drug design. Most tools rely on sequence-based statistical comparisons, such as information entropy of residue positions in aligned columns to infer location and properties of epitope sites. To facilitate cross-structural comparisons of epitopes on viral surface proteins, a python-based extraction tool implemented with Jupyter notebook is presented (Jupytope). Given a viral antigen structure of interest, a list of known epitope sites and a reference structure, the corresponding epitope structural properties can quickly be obtained. The tool integrates biopython modules for commonly used software such as NACCESS, DSSP as well as residue depth and outputs a list of structure-derived properties such as dihedral angles, solvent accessibility, residue depth and secondary structure that can be saved in several convenient data formats. To ensure correct spatial alignment, Jupytope takes a list of given epitope sites and their corresponding reference structure and aligns them before extracting the desired properties. Examples are demonstrated for epitopes of Influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) viral strains. The extracted properties assist detection of two Influenza subtypes and show potential in distinguishing between four major clades of SARS-CoV2, as compared with randomized labels. The tool will facilitate analytical and predictive works on viral epitopes through the extracted structural information. Jupytope and extracted datasets are available at https://github.com/shamimarashid/Jupytope.
Collapse
Affiliation(s)
- Shamima Rashid
- School of Computer Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Teng Ann Ng
- School of Computer Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Chee Keong Kwoh
- School of Computer Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| |
Collapse
|
12
|
Algorithmically-guided discovery of viral epitopes via linguistic parsing: Problem formulation and solving by soft computing. Appl Soft Comput 2022. [DOI: 10.1016/j.asoc.2022.109509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
13
|
Bioinformatics, Computational Informatics, and Modeling Approaches to the Design of mRNA COVID-19 Vaccine Candidates. COMPUTATION 2022. [DOI: 10.3390/computation10070117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This article is devoted to applying bioinformatics and immunoinformatics approaches for the development of a multi-epitope mRNA vaccine against the spike glycoproteins of circulating SARS-CoV-2 variants in selected African countries. The study’s relevance is dictated by the fact that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) began its global threat at the end of 2019 and since then has had a devastating impact on the whole world. Measures to reduce threats from the pandemic include social restrictions, restrictions on international travel, and vaccine development. In most cases, vaccine development depends on the spike glycoprotein, which serves as a medium for its entry into host cells. Although several variants of SARS-CoV-2 have emerged from mutations crossing continental boundaries, about 6000 delta variants have been reported along the coast of more than 20 countries in Africa, with South Africa accounting for the highest percentage. This also applies to the omicron variant of the SARS-CoV-2 virus in South Africa. The authors suggest that bioinformatics and immunoinformatics approaches be used to develop a multi-epitope mRNA vaccine against the spike glycoproteins of circulating SARS-CoV-2 variants in selected African countries. Various immunoinformatics tools have been used to predict T- and B-lymphocyte epitopes. The epitopes were further subjected to multiple evaluations to select epitopes that could elicit a sustained immunological response. The candidate vaccine consisted of seven epitopes, a highly immunogenic adjuvant, an MHC I-targeting domain (MITD), a signal peptide, and linkers. The molecular weight (MW) was predicted to be 223.1 kDa, well above the acceptable threshold of 110 kDa on an excellent vaccine candidate. In addition, the results showed that the candidate vaccine was antigenic, non-allergenic, non-toxic, thermostable, and hydrophilic. The vaccine candidate has good population coverage, with the highest range in East Africa (80.44%) followed by South Africa (77.23%). West Africa and North Africa have 76.65% and 76.13%, respectively, while Central Africa (75.64%) has minimal coverage. Among seven epitopes, no mutations were observed in 100 randomly selected SARS-CoV-2 spike glycoproteins in the study area. Evaluation of the secondary structure of the vaccine constructs revealed a stabilized structure showing 36.44% alpha-helices, 20.45% drawn filaments, and 33.38% random helices. Molecular docking of the TLR4 vaccine showed that the simulated vaccine has a high binding affinity for TLR-4, reflecting its ability to stimulate the innate and adaptive immune response.
Collapse
|
14
|
Khanum S, Carbone V, Gupta SK, Yeung J, Shu D, Wilson T, Parlane NA, Altermann E, Estein SM, Janssen PH, Wedlock DN, Heiser A. Mapping immunogenic epitopes of an adhesin-like protein from Methanobrevibacter ruminantium M1 and comparison of empirical data with in silico prediction methods. Sci Rep 2022; 12:10394. [PMID: 35729277 PMCID: PMC9213418 DOI: 10.1038/s41598-022-14545-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 06/08/2022] [Indexed: 11/09/2022] Open
Abstract
In silico prediction of epitopes is a potentially time-saving alternative to experimental epitope identification but is often subject to misidentification of epitopes and may not be useful for proteins from archaeal microorganisms. In this study, we mapped B- and T-cell epitopes of a model antigen from the methanogen Methanobrevibacter ruminantium M1, the Big_1 domain (AdLP-D1, amino acids 19-198) of an adhesin-like protein. A series of 17 overlapping 20-mer peptides was selected to cover the Big_1 domain. Peptide-specific antibodies were produced in mice and measured by ELISA, while an in vitro splenocyte re-stimulation assay determined specific T-cell responses. Overall, five peptides of the 17 peptides were shown to be major immunogenic epitopes of AdLP-D1. These immunogenic regions were examined for their localization in a homology-based model of AdLP-D1. Validated epitopes were found in the outside region of the protein, with loop like secondary structures reflecting their flexibility. The empirical data were compared with epitope predictions made by programmes based on a range of algorithms. In general, the epitopes identified by in silico predictions were not comparable to those determined empirically.
Collapse
Affiliation(s)
| | | | | | | | - Dairu Shu
- AgResearch, Palmerston North, New Zealand
| | | | | | - Eric Altermann
- AgResearch, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Silvia M Estein
- Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CONICET-CICPBA, Facultad de Ciencias Veterinarias, Campus Universitario, 7000, Tandil, Argentina
| | | | | | | |
Collapse
|
15
|
Naz S, Ahmad S, Abbasi SW, Ismail S, Waseem S, Tahir Ul Qamar M, Almatroudi A, Ali Z. Identification of immunodominant epitopes in allelic variants VK210 and VK247 of Plasmodium Vivax Circumsporozoite immunogen. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105120. [PMID: 34655808 DOI: 10.1016/j.meegid.2021.105120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 11/23/2022]
Abstract
Plasmodium vivax-induced malaria is among the leading causes of morbidity and mortality in sub-tropical and tropical regions and infect 2.85 billion people globally. The continual rise and propagation of resistance against anti-malarial drugs is a prerequisite to develop a potent vaccine candidate for Plasmodium vivax (P. vivax). Circumsporozoite protein (CSP) is an important immunogen of malaria parasite that has the conserved CSP structure as an immune dominant B-cell epitope. In current study, we focused on designing multi-epitope vaccines (MEVs) using various immunoinformatics tools against Pakistani based allelic variants VK210 and VK247 of P. vivax CSP (PvCSP) gene. Antigenicity, allergic potential and physicochemical parameters of both PvCSP variants were assessed for the designed MEVs and they were within acceptable range suitable for post experimental investigations. The three-dimensional structures of both MEVs have been predicted ab initio, optimized, and validated by using different online servers. The both MEVs candidates were stable and free from aggregation-prone regions. The stability of both MEVs had been improved by a disulfide engineering approach. To estimate the binding energy and stability of the MEVs, molecular docking simulation and binding free energy calculations with TLR-4 immune receptor have been conducted. The docking score of PvCSP210 and PvCSP247 for TLR-4 was -6.34 kJ/mol and - 2.3 kJ/mol, respectively. For PvCSP210-TLR4 system, mean RMSD was 4.96 Å while PvCSP247-TLR4 system, average RMSD was 4.49 Å. The binding free energy of PvCSP210-TLR4 complex and PvCSP247-TLR4 complex was -50.49/-117.15 kcal/mol (MMGBSA/MMPSA) and -52.94/-96.26 kcal/mol (MMGBSA/MMPSA), respectively. The expression of both MEVs produced in Escherichia coli K12 expression system by in silico cloning was significant. Immune simulation revealed that the proposed MEVs induce strong humoral and cellular immunological responses, in addition to significant production of interleukins and cytokines. In conclusions, we believed that the MEVs proposed in current research, using combine approach of immunoinformatics, structural biology and biophysical approaches, could induce protective and effective immune responses against P. vivax and the experimental validation of our findings could contribute to the development of potential malaria vaccine.
Collapse
Affiliation(s)
- Shumaila Naz
- NUMS Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Sumra Wajid Abbasi
- NUMS Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan.
| | - Saba Ismail
- NUMS Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Shahid Waseem
- Alpha Genomics (Pvt) Ltd., Plot 4-C, Danyal Plaza, Block A, Main Main PWD Rd, Sector A PWD Society, Islamabad
| | | | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Zain Ali
- Department of Biochemistry, Quaid-i-Azam University Islamabad, Pakistan
| |
Collapse
|
16
|
Bahai A, Asgari E, Mofrad MRK, Kloetgen A, McHardy AC. EpitopeVec: Linear Epitope Prediction Using Deep Protein Sequence Embeddings. Bioinformatics 2021; 37:4517-4525. [PMID: 34180989 PMCID: PMC8652027 DOI: 10.1093/bioinformatics/btab467] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/28/2021] [Accepted: 06/25/2021] [Indexed: 11/19/2022] Open
Abstract
Motivation B-cell epitopes (BCEs) play a pivotal role in the development of peptide vaccines, immuno-diagnostic reagents and antibody production, and thus in infectious disease prevention and diagnostics in general. Experimental methods used to determine BCEs are costly and time-consuming. Therefore, it is essential to develop computational methods for the rapid identification of BCEs. Although several computational methods have been developed for this task, generalizability is still a major concern, where cross-testing of the classifiers trained and tested on different datasets has revealed accuracies of 51–53%. Results We describe a new method called EpitopeVec, which uses a combination of residue properties, modified antigenicity scales, and protein language model-based representations (protein vectors) as features of peptides for linear BCE predictions. Extensive benchmarking of EpitopeVec and other state-of-the-art methods for linear BCE prediction on several large and small datasets, as well as cross-testing, demonstrated an improvement in the performance of EpitopeVec over other methods in terms of accuracy and area under the curve. As the predictive performance depended on the species origin of the respective antigens (viral, bacterial and eukaryotic), we also trained our method on a large viral dataset to create a dedicated linear viral BCE predictor with improved cross-testing performance. Availability and implementation The software is available at https://github.com/hzi-bifo/epitope-prediction. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Akash Bahai
- Computational Biology of Infection Research, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany.,Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig
| | - Ehsaneddin Asgari
- Computational Biology of Infection Research, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany.,Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Lab, Berkeley, CA 94720, USA
| | - Andreas Kloetgen
- Computational Biology of Infection Research, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
| | - Alice C McHardy
- Computational Biology of Infection Research, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany.,Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig
| |
Collapse
|
17
|
Olotu FA, Soliman MES. Immunoinformatics prediction of potential B-cell and T-cell epitopes as effective vaccine candidates for eliciting immunogenic responses against Epstein-Barr virus. Biomed J 2021; 44:317-337. [PMID: 34154948 PMCID: PMC8358216 DOI: 10.1016/j.bj.2020.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The ongoing search for viable treatment options to curtail Epstein Barr Virus (EBV) pathogenicity has necessitated a paradigmatic shift towards the design of peptide-based vaccines. Potential B-cell and T-cell epitopes were predicted for nine antigenic EBV proteins that mediate epithelial cell-attachment and spread, capsid self-assembly, DNA replication and processivity. METHODS Predictive algorithms incorporated in the Immune Epitope Database (IEDB) resources were used to determine potential B-cell epitopes based on their physicochemical attributes. These were combined with a string-kernel method and an antigenicity predictive AlgPred tool to enhance accuracy in the end-point selection of highly potential antigenic EBV B-cell epitopes. NetCTL 1.2 algorithms enabled the prediction of probable T-cell epitopes which were structurally modeled and subjected to blind peptide-protein docking with HLA-A*02:01. All-atom molecular dynamics (MD) simulation and Molecular Mechanics Generalized-Born Surface Area methods were used to investigate interaction dynamics and affinities of predicted T-cell peptide-protein complexes. RESULTS Computational predictions and sequence overlapping analysis yielded 18 linear (continuous) and discontinuous (conformational) subunit epitopes from the antigenic proteins with characteristic surface accessibility, flexibility and antigenicity, and predictive scores above the threshold value (1) set. A novel site was identified on HLA-A*02:01 with preferential affinity binding for modeled BMRF2, BXLF1 and BGLF4 T-cell epitopes. Interaction dynamics and energies were also computed in addition to crucial residues that mediated complex formation and stability. CONCLUSION This study implemented an integrative meta-analytical approach to model highly probable B-cell and T-cell epitopes as potential peptide-vaccine candidates for the treatment of EBV-related diseases.
Collapse
Affiliation(s)
- Fisayo A Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa.
| |
Collapse
|
18
|
Dar HA, Ismail S, Waheed Y, Ahmad S, Jamil Z, Aziz H, Hetta HF, Muhammad K. Designing a multi-epitope vaccine against Mycobacteroides abscessus by pangenome-reverse vaccinology. Sci Rep 2021; 11:11197. [PMID: 34045649 PMCID: PMC8159972 DOI: 10.1038/s41598-021-90868-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Mycobacteroides abscessus (Previously Mycobacterium abscessus) is an emerging microorganism of the newly defined genera Mycobacteroides that causes mainly skin and tissue diseases in humans. The recent availability of total 34 fully sequenced genomes of different strains belonging to this species has provided an opportunity to utilize this genomics data to gain novel insights and guide the development of specific antimicrobial therapies. In the present study, we collected collectively 34 complete genome sequences of M. abscessus from the NCBI GenBank database. Pangenome analysis was conducted on these genomes to understand the genetic diversity and to obtain proteins associated with its core genome. These core proteins were then subjected to various subtractive filters to identify potential antigenic targets that were subjected to multi-epitope vaccine design. Our analysis projected the open pangenome of M. abscessus containing 3443 core genes. After applying various stepwise filtration steps on the core proteins, a total of four potential antigenic targets were identified. Utilizing their constituent CD4 and CD8 T-cell epitopes, a multi-epitope based subunit vaccine was computationally designed. Sequence-based analysis as well as structural characterization revealed the immunological effectiveness of this designed vaccine. Further molecular docking, molecular dynamics simulation and binding free energy estimation with Toll-like receptor 2 indicated strong structural associations of the vaccine with the immune receptor. The promising results are encouraging and need to be validated by additional wet laboratory studies for confirmation.
Collapse
Affiliation(s)
- Hamza Arshad Dar
- Foundation University Medical College, Foundation University Islamabad, DHA-I, Islamabad, 44000, Pakistan
| | - Saba Ismail
- Foundation University Medical College, Foundation University Islamabad, DHA-I, Islamabad, 44000, Pakistan
| | - Yasir Waheed
- Foundation University Medical College, Foundation University Islamabad, DHA-I, Islamabad, 44000, Pakistan.
| | - Sajjad Ahmad
- Foundation University Medical College, Foundation University Islamabad, DHA-I, Islamabad, 44000, Pakistan
| | - Zubia Jamil
- Foundation University Medical College, Foundation University Islamabad, DHA-I, Islamabad, 44000, Pakistan
| | - Hafsa Aziz
- Nuclear Medicine, Oncology, and Radiotherapy Institute, Islamabad, 44000, Pakistan
| | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, 15551, Al Ain, United Arab Emirates.
| |
Collapse
|
19
|
Chizari M, Fani-Kheshti S, Taeb J, Farajollahi MM, Mohsenzadegan M. The Anti-Proliferative Effect of a Newly-Produced Anti-PSCA-Peptide Antibody by Multiple Bioinformatics Tools, on Prostate Cancer Cells. Recent Pat Anticancer Drug Discov 2021; 16:73-83. [PMID: 33176663 DOI: 10.2174/1574892815999201110212411] [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] [Received: 06/12/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prostate Stem Cell Antigen (PSCA) is a small cell surface protein, overexpressed in 90% of prostate cancers. Determination of epitopes that elicit an appropriate response to the antibody generation is vital for diagnostic and immunotherapeutic purposes for prostate cancer treatment. Presently, bioinformatics B-cell prediction tools can predict the location of epitopes, which is uncomplicated, faster, and more cost-effective than experimental methods. OBJECTIVE We aimed to predict a novel linear peptide for Prostate Stem Cell Antigen (PSCA) protein in order to generate anti-PSCA-peptide (p) antibody and to investigate its effect on prostate cancer cells. METHODS In the current study, a novel linear peptide for PSCA was predicted using in silico methods that utilize a set of linear B-cell epitope prediction tools. Polyclonal antibody (anti-PSCA-p antibody "Patent No. 99318") against PSCA peptide was generated. The antibody reactivity was determined by the Enzyme-Linked Immunosorbent Assay (ELISA) and its specificity by immunocytochemistry (ICC), immunohistochemistry (IHC), and Western Blotting (WB) assays. The effect of the anti-PSCA-p antibody on PSCA-expressing prostate cancer cell line was assessed by Methylthiazolyldiphenyl- Tetrazolium bromide (MTT) assay. RESULTS New peptide-fragment of PSCA sequence as "N-CVDDSQDYYVGKKN-C" (PSCA-p) was selected and synthesized. The anti-PSCA-p antibody against the PSCA-p showed immunoreactivity with PSCA-p specifically bound to PC-3 cells. Also, the anti-PSCA-p antibody strongly stained the prostate cancer tissues as compared to Benign Prostatic Hyperplasia (BPH) and normal tissues (P < 0.001). As the degree of malignancy increased, the staining intensity was also elevated in prostate cancer tissue (P < 0.001). Interestingly, the anti-PSCA-p antibody showed anti-proliferative effects on PC-3 cells (31%) with no growth inhibition effect on PSCA-negative cells. CONCLUSION In this study, we developed a new peptide sequence (PSCA-p) of PSCA. The PSCA-p targeting by anti-PSCA-p antibody inhibited the proliferation of prostate cancer cells, suggesting the potential of PSCA-p immunotherapy for future prostate cancer studies.
Collapse
Affiliation(s)
- Milad Chizari
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Fani-Kheshti
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Jaleh Taeb
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad M Farajollahi
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Monireh Mohsenzadegan
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Hasan MM, Khatun MS, Kurata H. iLBE for Computational Identification of Linear B-cell Epitopes by Integrating Sequence and Evolutionary Features. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:593-600. [PMID: 33099033 PMCID: PMC8377379 DOI: 10.1016/j.gpb.2019.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/13/2019] [Accepted: 04/19/2019] [Indexed: 12/17/2022]
Abstract
Linear B-cell epitopes are critically important for immunological applications, such as vaccine design, immunodiagnostic test, and antibody production, as well as disease diagnosis and therapy. The accurate identification of linear B-cell epitopes remains challenging despite several decades of research. In this work, we have developed a novel predictor, Identification of Linear B-cell Epitope (iLBE), by integrating evolutionary and sequence-based features. The successive feature vectors were optimized by a Wilcoxon-rank sum test. Then the random forest (RF) algorithm using the optimal consecutive feature vectors was applied to predict linear B-cell epitopes. We combined the RF scores by the logistic regression to enhance the prediction accuracy. iLBE yielded an area under curve score of 0.809 on the training dataset and outperformed other prediction models on a comprehensive independent dataset. iLBE is a powerful computational tool to identify the linear B-cell epitopes and would help to develop penetrating diagnostic tests. A web application with curated datasets for iLBE is freely accessible at http://kurata14.bio.kyutech.ac.jp/iLBE/.
Collapse
Affiliation(s)
- Md Mehedi Hasan
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Mst Shamima Khatun
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan; Biomedical Informatics R&D Center, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan.
| |
Collapse
|
21
|
Designing a multi-epitope peptide based vaccine against SARS-CoV-2. Sci Rep 2020; 10:16219. [PMID: 33004978 PMCID: PMC7530768 DOI: 10.1038/s41598-020-73371-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
COVID-19 pandemic has resulted in 16,114,449 cases with 646,641 deaths from the 217 countries, or territories as on July 27th 2020. Due to multifaceted issues and challenges in the implementation of the safety and preventive measures, inconsistent coordination between societies-governments and most importantly lack of specific vaccine to SARS-CoV-2, the spread of the virus that initially emerged at Wuhan is still uprising after taking a heavy toll on human life. In the present study, we mapped immunogenic epitopes present on the four structural proteins of SARS-CoV-2 and we designed a multi-epitope peptide based vaccine that, demonstrated a high immunogenic response with a vast application on world’s human population. On codon optimization and in-silico cloning, we found that candidate vaccine showed high expression in E. coli and immune simulation resulted in inducing a high level of both B-cell and T-cell mediated immunity. The results predicted that exposure of vaccine by administrating three injections significantly subsidized the antigen growth in the system. The proposed candidate vaccine found promising by yielding desired results and hence, should be validated by practical experimentations for its functioning and efficacy to neutralize SARS-CoV-2.
Collapse
|
22
|
Kaur H, Shorie M, Sabherwal P. Biolayer interferometry-SELEX for Shiga toxin antigenic-peptide aptamers & detection via chitosan-WSe 2 aptasensor. Biosens Bioelectron 2020; 167:112498. [PMID: 32814208 DOI: 10.1016/j.bios.2020.112498] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022]
Abstract
We report biolayer interferometry based in-vitro selection technique (BLI-SELEX) for fishing out specific aptamers against E. coli Shiga toxin subtypes viz., stx1 & stx2 via epitopic peptides. BLI-SELEX is a one-step technique for rapidly generating aptamers against protein biomarkers in a microtiter plate format, obliterating the need for multiple enrichment rounds to harvest high-affinity aptamers as in conventional SELEX. Two unique aptamers selected against stx1 & stx2 with picomolar Kd (~47 pM & ~29 pM, respectively) were successfully used to fabricate voltammetric diagnostic assay via immobilization onto chitosan exfoliated 2D tungsten diselenide (WSe2) nanosheet platform. These aptamers modified nanosensors showed high sensitivity of ~ 5.0 μA ng-1 mL, a dynamic response range from 50 pg mL-1 to 100 ng mL-1, with a detection limit of 44.5 pg mL-1 & 41.3 pg mL-1 for stx subtypes, respectively and showed low cross-reactivity in spiked urine, serum and milk samples. The synergistic effect of selective aptamers & high sensitivity imparted by 2D transition metal dichalcogenide (TMD) highlights the superior potential of a fabricated nanosensor for bacterial toxin detection.
Collapse
Affiliation(s)
- Harmanjit Kaur
- Institute of Nano Science & Technology, Mohali, 160062, India
| | - Munish Shorie
- Institute of Nano Science & Technology, Mohali, 160062, India
| | | |
Collapse
|
23
|
Kaumaya PTP. B-cell epitope peptide cancer vaccines: a new paradigm for combination immunotherapies with novel checkpoint peptide vaccine. Future Oncol 2020; 16:1767-1791. [PMID: 32564612 PMCID: PMC7426751 DOI: 10.2217/fon-2020-0224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
In light of the numerous US FDA-approved humanized monoclonal antibodies (mAbs) for cancer immunotherapy, it is surprising that the advancement of B-cell epitope vaccines designed to elicit a natural humoral polyclonal antibody response has not gained traction in the immune-oncology landscape. Passive immunotherapy with humanized mAbs (Trastuzumab [Herceptin®]; Pertuzumab [Perjeta®]) has provided clinical benefit to breast cancer patients, albeit with significant shortcomings including toxicity problems and resistance, high costs, sophisticated therapeutic regimen and long half-life. The role of B-cell humoral immunity in cancer is under appreciated and underdeveloped. We have advanced the idea of active immunotherapy with chimeric B-cell epitope peptides incorporating a 'promiscuous' T-cell epitope that elicits a polyclonal antibody response, which provides safe, cost-effective therapeutic advantage over mAbs. We have created a portfolio of validated B-cell peptide epitopes against multiple receptor tyrosine kinases (HER-1, HER-3, IGF-1R and VEGF). We have successfully translated two HER-2 combination B-cell peptide vaccines in Phase I and II clinical trials. We have recently developed an effective novel PD-1 vaccine. In this article, I will review our approaches and strategies that focus on B-cell epitope cancer vaccines.
Collapse
Affiliation(s)
- Pravin TP Kaumaya
- Department of Obstetrics & Gynecology, College of Medicine, Wexner Medical Center, The James Cancer Hospital & Solove Research Institute, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Anwar S, Mourosi JT, Khan MF, Hosen MJ. Prediction of Epitope-Based Peptide Vaccine Against the Chikungunya Virus by Immuno-informatics Approach. Curr Pharm Biotechnol 2020; 21:325-340. [PMID: 31721709 DOI: 10.2174/1389201020666191112161743] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/16/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chikungunya is an arthropod-borne viral disease characterized by abrupt onset of fever frequently accompanied by joint pain, which has been identified in over 60 countries in Africa, the Americas, Asia, and Europe. METHODS Regardless of the availability of molecular knowledge of this virus, no definite vaccine or other remedial agents have been developed yet. In the present study, a combination of B-cell and T-cell epitope predictions, followed by molecular docking simulation approach has been carried out to design a potential epitope-based peptide vaccine, which can trigger a critical immune response against the viral infections. RESULTS A total of 52 sequences of E1 glycoprotein from the previously reported isolates of Chikungunya outbreaks were retrieved and examined through in silico methods to identify a potential B-cell and T-cell epitope. From the two separate epitope prediction servers, five potential B-cell epitopes were selected, among them "NTQLSEAHVEKS" was found highly conserved across strains and manifests high antigenicity with surface accessibility, flexibility, and hydrophilicity. Similarly, two highly conserved, non-allergenic, non-cytotoxic putative T-cell epitopes having maximum population coverage were screened to bind with the HLA-C 12*03 molecule. Molecular docking simulation revealed potential T-cell based epitope "KTEFASAYR" as a vaccine candidate for this virus. CONCLUSION A combination of these B-cell and T-cell epitope-based vaccine can open up a new skyline with broader therapeutic application against Chikungunya virus with further experimental and clinical investigation.
Collapse
Affiliation(s)
- Saeed Anwar
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh.,Maternal and Child Health Program, Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 8440 112 St. NW, Edmonton, AB T6G 2R7, Canada
| | - Jarin T Mourosi
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh.,Microbial and Cellular Biology Program, Department of Biology, The Catholic University of America, 620 Michigan Ave. NE, Washington, DC, 20064, United States
| | - Md Fahim Khan
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Mohammad J Hosen
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| |
Collapse
|