1
|
Shen Y, Jiang Z, Liu R. Dynamic integration of feature- and template-based methods improves the prediction of conformational B cell epitopes. Structure 2025; 33:798-807.e4. [PMID: 39938510 DOI: 10.1016/j.str.2025.01.018] [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: 10/22/2024] [Revised: 12/10/2024] [Accepted: 01/16/2025] [Indexed: 02/14/2025]
Abstract
The accurate prediction of conformational epitopes promotes our understanding of antigen-antibody interactions. All existing algorithms depend on a feature-based strategy, which limits their performance. A template-based strategy can provide complementary information, and the interplay between these two strategies could improve the prediction of epitopes. Here, we present DynaBCE, a dynamic ensemble algorithm to effectively identify conformational B cell epitopes (BCEs). Using novel handcrafted structural descriptors and embeddings from protein language models, we developed machine learning and deep learning modules based on boosting algorithms and geometric graph neural networks, respectively. Furthermore, we built a template module by leveraging known structural template information and transformer-based algorithms to capture binding signatures. Finally, we integrated the three modules using a dynamic weighting approach to maximize the strength of each module for different samples. DynaBCE achieved promising results for both native and predicted structures and outperformed previous methods as demonstrated in various evaluation scenarios.
Collapse
Affiliation(s)
- Yueyue Shen
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Zheng Jiang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Rong Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China.
| |
Collapse
|
2
|
Yang SQ, Yang K, Li XR, Zheng Y, Cao SJ, Yan QG, Huang XB, Wen YP, Zhao Q, Du SY, Lang YF, Zhao S, Li C, Wu R. A novel linear B cell epitope of the porcine circovirus type 3 capsid protein identified by phage display technology. J Virol Methods 2025; 333:115080. [PMID: 39638262 DOI: 10.1016/j.jviromet.2024.115080] [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: 09/17/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Porcine circovirus type 3 (PCV3) is endemic in swine worldwide and causes reproductive disorders, dermatitis and nephrotic syndrome, and multi-organ inflammation. PCV3 capsid protein (Cap) can self-assemble into virus-like particles (VLPs), and is an ideal candidate for vaccines and diagnostic reagents.In this study, the recombinant PCV3 Cap protein was successfully expressed in E. coli by deleting the nuclear localization sequence (NLS). The PCV3 VLPs were observed by transmission electron microscopy, and its immunogenicity was evaluated in six-week-old female BALB/c mice. A monoclonal antibody was named mAb 2D6, and demonstrated strong reactivity and specificity to PCV3 Cap. The purified mAb 2D6 was further used for bio-panning to select phage expressing specific epitopes from phage-displayed 7 mer-peptide library. A novel linear B-cell epitope, recognized by mAb 2D6, was identified at the amino acid region 47-53 of Cap. The phage peptide sequences were analyzed using multiple sequence alignment and evaluated by peptide ELISA. These results provide insights for developing diagnostic tools and potential vaccines for PCV3.
Collapse
Affiliation(s)
- Shu-Qing Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ke Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin-Ran Li
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi Zheng
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - San-Jie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qi-Gui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Bo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Ping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sen-Yan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Fei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chun Li
- Sichuan Animal Disease Prevention and Control Center, Chengdu 610047, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; National Animal Experiments Teaching Demonstration Center, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
3
|
Souffou M, Dechavanne C, Kammoun Z, Viwami F, Gaugué I, Beldjoudi N, Dechavanne S, Sare N, Garcia A, Dambrun M, Migot-Nabias F. Functionality of Toxoplasma gondii antibodies in a population of Beninese pregnant women exposed to malaria. Sci Rep 2025; 15:9303. [PMID: 40102442 PMCID: PMC11920409 DOI: 10.1038/s41598-025-91803-5] [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: 10/24/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Plasmodium falciparum and Toxoplasma gondii are two apicomplexan parasites that can lead to severe complications for the newborn when contracted during pregnancy. This study explores the cross-reactivity of antibodies specific to both pathogens in pregnant women, exposed or not to malaria. The antibody response against full-length recombinant antigens from P. falciparum (PfAMA1, Pfs48/45) and T. gondii (TgAMA1, TgSAG1, TgGRA7), selected for their strong immunogenicity, was analysed on 150 plasma samples from women residing in Benin or France. The antibody functionality was assessed using P. falciparum in vitro Growth Inhibition Assay (GIA). As the main results, toxoplasmosis seropositive women with an ongoing P. falciparum infection better inhibited P. falciparum invasion compared to toxoplasmosis seronegative women (34.6% vs. 17.2%, p ≤ 0.01). Women with positive serologies for both parasites presented a significantly higher inhibition of P. falciparum invasion compared to those only seropositive for malaria (coef = 6.27, p = 0.076) in reference with double-negative women (coef = 11.35, p = 0.001). These data suggest that plasma samples containing anti-T. gondii IgG may contribute reducing the development of P. falciparum parasites. This study provides insight into the immune dynamics of the co-infection by these two apicomplexans with potential implications for developing cross-protective vaccines and therapies.
Collapse
Grants
- PhD scholarship Conseil Départemental de Mayotte
- IdEx 2019 "Dynamique Recherche", PlasDCty Université Paris Cité, France
- DHU 'Risks and pregnancy", PRIDE 2016, TOXODIAG AP-HP Nord et Université Paris Cité
- DHU 'Risks and pregnancy", PRIDE 2016, TOXODIAG AP-HP Nord et Université Paris Cité
- grant 0602DIRmba, 2018, CoaLa Institut de Médecine et d'Epidémiologie Appliquée
- grant 0602DIRmba, 2018, CoaLa Institut de Médecine et d'Epidémiologie Appliquée
- ANR-19-CE44-0004, IgName Agence Nationale de la Recherche
Collapse
Affiliation(s)
- Mariama Souffou
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Célia Dechavanne
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Zaineb Kammoun
- Université Paris Cité, Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), CNRS, Paris, France
| | - Firmine Viwami
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Isabelle Gaugué
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
- Genetics and Developmental Biology, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Paris, 75005, France
| | - Naima Beldjoudi
- Epidemiology and Clinical Research Department, GH Paris Nord Val de Seine, Assistance Publique - Hôpitaux de Paris, Paris, France
| | | | - Nawal Sare
- Centre d'Etude et de Recherche sur les Pathologies Associées à la Grossesse et à L'Enfance, Cotonou, Bénin
| | - André Garcia
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Magalie Dambrun
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France.
| | | |
Collapse
|
4
|
Liu M, Ren T, Zhang L, Li P, Zhong Z, Zhou L, Qin Y, Ouyang K, Chen Y, Huang W, Wei Z. Development of a monoclonal antibody specifically recognizing a linear epitope on the E1 protein of Getah virus. Virology 2025; 602:110315. [PMID: 39626608 DOI: 10.1016/j.virol.2024.110315] [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: 07/31/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024]
Abstract
Getah virus (GETV) is a mosquito-borne alphavirus that can cause disease outbreaks in domesticated mammals. The E1 protein of GETV plays a crucial role in mediating the fusion of viruses and host cells. In this study, the recombinant GETV E1 protein was expressed and administered to BALB/c mice. Hybridoma cells secreting a monoclonal antibody (mAb) 7D4-2 against E1 protein were subsequently obtained using a cell fusion protocol between SP2/0 cells and splenocytes. The reactivity of mAb 7D4-2 with GETV was confirmed through Western blot analysis and indirect immunofluorescence assay (IFA). A precise B cell linear epitope, 281-DIPDTAF-287, was identified using Western blot analysis with the produced mAb 7D4-2 by constructing and expressing a series of truncated His-fused E1 proteins. Conservation analysis indicated that this epitope is highly conserved in Group III strains of GETV, but exhibits an amino acid difference (T285S) compared to Group I, Group II, and Group IV. Cross-reactivity analysis by Western blot demonstrated that the B-cell epitope containing the mutation could be recognized by mAb 7D4-2. The ability of mAb 7D4-2 to recognize the epitope carrying the T285S mutation was further confirmed using an infectious clone of GETV. This study enhances the understanding of the biological characteristics of the E1 protein and will facilitate the future development of diagnostic tests for GETV.
Collapse
Affiliation(s)
- Muyang Liu
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Tongwei Ren
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Liping Zhang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Peijie Li
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Zhen Zhong
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Lingshan Zhou
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Yifeng Qin
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530005, China; Guangxi Key Laboratory of Animal Breeding and Disease Control and Prevention, Nanning, 530005, China
| | - Kang Ouyang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530005, China; Guangxi Key Laboratory of Animal Breeding and Disease Control and Prevention, Nanning, 530005, China
| | - Ying Chen
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530005, China; Guangxi Key Laboratory of Animal Breeding and Disease Control and Prevention, Nanning, 530005, China
| | - Weijian Huang
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530005, China; Guangxi Key Laboratory of Animal Breeding and Disease Control and Prevention, Nanning, 530005, China
| | - Zuzhang Wei
- Laboratory of Animal Infectious Diseases and Molecular Immunology, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530005, China; Guangxi Key Laboratory of Animal Breeding and Disease Control and Prevention, Nanning, 530005, China.
| |
Collapse
|
5
|
Ottobre M, Van Snick J, Aparicio JL. Interleukin-17A (IL-17A) is involved in antibody specificity to conformational epitopes. Biochem Biophys Res Commun 2024; 739:150588. [PMID: 39191146 DOI: 10.1016/j.bbrc.2024.150588] [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: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The specificity of antibodies (Ab) is essential for the precise recognition of foreign or dangerous molecules. We have shown that mice infected with non-pathogenic Lactate Dehydrogenase Elevating Virus (LDV) inoculated with human growth hormone (hGH) or Ovalbumin (OVA), exhibit modified specificity of anti-hGH or anti-OVA Ab associated with the secretion of IFN-γ, IL-13, and IL-17. Cytokines are directly or indirectly involved in the isotypes, specificity, and affinity of Ab. Accordingly, here we investigated the effect of IL-17 neutralization on Ab specificities to OVA and Diphtheria Toxoid (DTx) in a mouse model of viral infection. Thereby, we employed an anti-cytokine "auto-vaccination" with an OVA/IL-17A complex or a Monoclonal Ab (MAb) anti-IL-17A (MM17/F3). Competitive ELISA assays were used to estimate the quality of the humoral immune response and the amount of Abs to conformational versus linear antigenic determinants. Results indicated that the OVA/IL-17A complex increased Abs levels to conformational epitopes of OVA, while LDV prolonged antibodies for a longer period. Mice treated with MM17F3 MAb showed an increase in Abs to conformational epitopes of OVA. A similar effect, confirmed by a competitive Western-blot assay, was produced by LDV. Moreover, an increased level of IgM, IgG1, and IgG2a was found in infected animals. Similarly, MAb anti-IL-17A treatment increased the proportion of Ab to conformational epitopes of DTx in uninfected mice, while LDV decreased this parameter. In conclusion, our findings demonstrate a correlation between IL-17A neutralization and a change in Ab specificity to OVA or DTx, presenting a novel strategy for obtaining Abs with higher specificity.
Collapse
Affiliation(s)
- Macarena Ottobre
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Jacques Van Snick
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
| | - José L Aparicio
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Madsen AV, Mejias-Gomez O, Pedersen LE, Preben Morth J, Kristensen P, Jenkins TP, Goletz S. Structural trends in antibody-antigen binding interfaces: a computational analysis of 1833 experimentally determined 3D structures. Comput Struct Biotechnol J 2024; 23:199-211. [PMID: 38161735 PMCID: PMC10755492 DOI: 10.1016/j.csbj.2023.11.056] [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: 10/08/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Antibodies are attractive therapeutic candidates due to their ability to bind cognate antigens with high affinity and specificity. Still, the underlying molecular rules governing the antibody-antigen interface remain poorly understood, making in silico antibody design inherently difficult and keeping the discovery and design of novel antibodies a costly and laborious process. This study investigates the characteristics of antibody-antigen binding interfaces through a computational analysis of more than 850,000 atom-atom contacts from the largest reported set of antibody-antigen complexes with 1833 nonredundant, experimentally determined structures. The analysis compares binding characteristics of conventional antibodies and single-domain antibodies (sdAbs) targeting both protein- and peptide antigens. We find clear patterns in the number antibody-antigen contacts and amino acid frequencies in the paratope. The direct comparison of sdAbs and conventional antibodies helps elucidate the mechanisms employed by sdAbs to compensate for their smaller size and the fact that they harbor only half the number of complementarity-determining regions compared to conventional antibodies. Furthermore, we pinpoint antibody interface hotspot residues that are often found at the binding interface and the amino acid frequencies at these positions. These findings have direct potential applications in antibody engineering and the design of improved antibody libraries.
Collapse
Affiliation(s)
- Andreas V. Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Oscar Mejias-Gomez
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Lasse E. Pedersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - J. Preben Morth
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Peter Kristensen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Timothy P. Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
7
|
Kang W, Zhang J, Yu N, Wei L, Chen Y. Screening of IgE-Binding Epitopes of Peach Allergenic Protein Pru p 7 Based on an Immune Microarray Assay. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23991-24002. [PMID: 39422561 DOI: 10.1021/acs.jafc.4c00347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Pru p 7 (also named Peamaclein) is a member of the gibberellin-regulated protein family, which is the latest foodborne allergenic protein identified in peach. In this paper, the prokaryotic expression and identification of Pru p 7 were performed, and the protein properties, structure, and homology were analyzed. In addition, a preliminary screening of B-cell linear epitopes of Pru p 7 was performed by the bioinformatics software prediction method, and three epitopes were identified using slot-blot immune microarray assay combined with an immune score matrix (P-1, AA16-21, AGYQER; P-2, AA40-46, TYGNKDE; P-3, AA52-59, DLKNSKGN). Moreover, the electrostatic potential of these epitopes was analyzed, and the stability after ultrahigh pressure treatment was also verified. Finally, the amino acids that play key immune roles in the epitopes were obtained by amino acid mutations. These results may contribute to the further understanding of Pru p 7 and the prevention of peach allergy.
Collapse
Affiliation(s)
- Wenhan Kang
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Jiukai Zhang
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Ning Yu
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Liyang Wei
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Ying Chen
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| |
Collapse
|
8
|
Xin C, Chen Z, Zhou J, Chen Y, Liu Y, Liu H, Liang C, Zhu X, Qi Y, Zhang G, Wang A. Identification and characterization of linear B-cell epitopes on African swine fever virus H171R protein. Microbiol Spectr 2024; 12:e0141124. [PMID: 39440988 PMCID: PMC11619302 DOI: 10.1128/spectrum.01411-24] [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: 06/10/2024] [Accepted: 08/03/2024] [Indexed: 10/25/2024] Open
Abstract
African swine fever virus (ASFV) is a highly contagious and often fatal pathogen that poses a significant threat to the swine industry worldwide. The H171R protein, a structural component of ASFV, plays crucial roles in viral assembly, host cell entry, and modulation of the host immune response. This study aimed to comprehensively characterize the linear B-cell epitopes on the H171R protein to facilitate the development of diagnostic tools and subunit vaccines against ASFV. A combined approach involving bioinformatics analysis and experimental techniques was employed. The recombinant H171R protein was expressed and purified, and specific monoclonal antibodies were generated through immunization and hybridoma technology. Systematic epitope mapping using overlapping peptide fragments and alanine-scanning mutagenesis revealed four minimal linear epitopes: 84HPLLPYQQSSDEQP97, 93SDEQPMMPYQQPPG106, 111PYEQIYHKKHASQQ124, and 129LNDYYQHILALGDED143. The identified epitopes exhibited strong immunogenicity, as demonstrated by their reactivity with ASFV-positive swine sera. Critical amino acid residues within each epitope were identified through mutational analysis. Structural modeling and visualization of the H171R protein provided insights into the spatial distribution and accessibility of the epitope regions. These findings contribute to a better understanding of the H171R protein's antigenic properties and lay the foundation for developing effective diagnostic assays and subunit vaccines against African swine fever. IMPORTANCE African swine fever virus (ASFV) poses a severe threat to the global swine industry. This study characterizes linear B-cell epitopes on the crucial ASFV H171R protein, facilitating the development of improved diagnostics and subunit vaccines. Four immunogenic epitopes were identified, offering valuable information for designing sensitive diagnostic assays and potential subunit vaccine candidates. By advancing the understanding of H171R's antigenic landscape, this research contributes to controlling ASFV's devastating impacts, safeguarding the swine industry, and ensuring food security.
Collapse
Affiliation(s)
- Cheng Xin
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Zhuting Chen
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Jingming Zhou
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Yankai Liu
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Hongliang Liu
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Chao Liang
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Xifang Zhu
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Yanhua Qi
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| | - Gaiping Zhang
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
- School of Advanced Agricultural Sciences, Peking University, Beijing, China
- College of Veterinary Medicine, Henan Agricultural University, Henan, Zhengzhou, China
| | - Aiping Wang
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou, China
- Longhu Laboratory, Henan, Zhengzhou, China
- Henan Key Laboratory of Immunobiology, Henan, Zhengzhou, China
| |
Collapse
|
9
|
Carroll M, Rosenbaum E, Viswanathan R. Computational Methods to Predict Conformational B-Cell Epitopes. Biomolecules 2024; 14:983. [PMID: 39199371 PMCID: PMC11352882 DOI: 10.3390/biom14080983] [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: 07/09/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
Accurate computational prediction of B-cell epitopes can greatly enhance biomedical research and rapidly advance efforts to develop therapeutics, monoclonal antibodies, vaccines, and immunodiagnostic reagents. Previous research efforts have primarily focused on the development of computational methods to predict linear epitopes rather than conformational epitopes; however, the latter is much more biologically predominant. Several conformational B-cell epitope prediction methods have recently been published, but their predictive performances are weak. Here, we present a review of the latest computational methods and assess their performances on a diverse test set of 29 non-redundant unbound antigen structures. Our results demonstrate that ISPIPab performs better than most methods and compares favorably with other recent antigen-specific methods. Finally, we suggest new strategies and opportunities to improve computational predictions of conformational B-cell epitopes.
Collapse
Affiliation(s)
| | | | - R. Viswanathan
- Department of Chemistry and Biochemistry, Yeshiva College, Yeshiva University, New York, NY 10033, USA; (M.C.); (E.R.)
| |
Collapse
|
10
|
Rogers J, Bajur AT, Salaita K, Spillane KM. Mechanical control of antigen detection and discrimination by T and B cell receptors. Biophys J 2024; 123:2234-2255. [PMID: 38794795 PMCID: PMC11331051 DOI: 10.1016/j.bpj.2024.05.020] [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: 02/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The adaptive immune response is orchestrated by just two cell types, T cells and B cells. Both cells possess the remarkable ability to recognize virtually any antigen through their respective antigen receptors-the T cell receptor (TCR) and B cell receptor (BCR). Despite extensive investigations into the biochemical signaling events triggered by antigen recognition in these cells, our ability to predict or control the outcome of T and B cell activation remains elusive. This challenge is compounded by the sensitivity of T and B cells to the biophysical properties of antigens and the cells presenting them-a phenomenon we are just beginning to understand. Recent insights underscore the central role of mechanical forces in this process, governing the conformation, signaling activity, and spatial organization of TCRs and BCRs within the cell membrane, ultimately eliciting distinct cellular responses. Traditionally, T cells and B cells have been studied independently, with researchers working in parallel to decipher the mechanisms of activation. While these investigations have unveiled many overlaps in how these cell types sense and respond to antigens, notable differences exist. To fully grasp their biology and harness it for therapeutic purposes, these distinctions must be considered. This review compares and contrasts the TCR and BCR, placing emphasis on the role of mechanical force in regulating the activity of both receptors to shape cellular and humoral adaptive immune responses.
Collapse
Affiliation(s)
- Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom; Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
11
|
Raposo B, Klareskog L, Robinson WH, Malmström V, Grönwall C. The peculiar features, diversity and impact of citrulline-reactive autoantibodies. Nat Rev Rheumatol 2024; 20:399-416. [PMID: 38858604 DOI: 10.1038/s41584-024-01124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
Since entering the stage 25 years ago as a highly specific serological biomarker for rheumatoid arthritis, anti-citrullinated protein antibodies (ACPAs) have been a topic of extensive research. This hallmark B cell response arises years before disease onset, displays interpatient autoantigen variability, and is associated with poor clinical outcomes. Technological and scientific advances have revealed broad clonal diversity and intriguing features including high levels of somatic hypermutation, variable-domain N-linked glycosylation, hapten-like peptide interactions, and clone-specific multireactivity to citrullinated, carbamylated and acetylated epitopes. ACPAs have been found in different isotypes and subclasses, in both circulation and tissue, and are secreted by both plasmablasts and long-lived plasma cells. Notably, although some disease-promoting features have been reported, results now demonstrate that certain monoclonal ACPAs therapeutically block arthritis and inflammation in mouse models. A wealth of functional studies using patient-derived polyclonal and monoclonal antibodies have provided evidence for pathogenic and protective effects of ACPAs in the context of arthritis. To understand the roles of ACPAs, one needs to consider their immunological properties by incorporating different facets such as rheumatoid arthritis B cell biology, environmental triggers and chronic antigen exposure. The emerging picture points to a complex role of citrulline-reactive autoantibodies, in which the diversity and dynamics of antibody clones could determine clinical progression and manifestations.
Collapse
Affiliation(s)
- Bruno Raposo
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Vivianne Malmström
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Ras-Carmona A, Reche PA. Analysis of Virus-Specific B Cell Epitopes Reveals Extensive Antigen Degradation Prior to Recognition. Cells 2024; 13:1076. [PMID: 38994930 PMCID: PMC11240346 DOI: 10.3390/cells13131076] [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: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024] Open
Abstract
B cell epitopes must be visible for recognition by cognate B cells and/or antibodies. Here, we studied that premise for known linear B cell epitopes that were collected from the Immune Epitope Database as being recognized by humans during microbial infections. We found that the majority of such known B cell epitopes are virus-specific linear B cell epitopes (87.96%), and most are located in antigens that remain enclosed in host cells and/or virus particles, preventing antibody recognition (18,832 out of 29,225 epitopes). Moreover, we estimated that only a minority (32.72%) of the virus-specific linear B cell epitopes that are found in exposed viral regions (e.g., the ectodomains of envelope proteins) are solvent accessible on intact antigens. Hence, we conclude that ample degradation/processing of viral particles and/or infected cells must occur prior to B cell recognition, thus shaping the B cell epitope repertoire.
Collapse
Affiliation(s)
| | - Pedro A. Reche
- Laboratory of Immunomedicine, Department of Immunology & O2, Faculty of Medicine, University Complutense of Madrid, Pza Ramon y Cajal S/N, 28040 Madrid, Spain;
| |
Collapse
|
13
|
Rahman S, Chiou CC, Ahmad S, Islam ZU, Tanaka T, Alouffi A, Chen CC, Almutairi MM, Ali A. Subtractive Proteomics and Reverse-Vaccinology Approaches for Novel Drug Target Identification and Chimeric Vaccine Development against Bartonella henselae Strain Houston-1. Bioengineering (Basel) 2024; 11:505. [PMID: 38790371 PMCID: PMC11118080 DOI: 10.3390/bioengineering11050505] [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: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Bartonella henselae is a Gram-negative bacterium causing a variety of clinical symptoms, ranging from cat-scratch disease to severe systemic infections, and it is primarily transmitted by infected fleas. Its status as an emerging zoonotic pathogen and its capacity to persist within host erythrocytes and endothelial cells emphasize its clinical significance. Despite progress in understanding its pathogenesis, limited knowledge exists about the virulence factors and regulatory mechanisms specific to the B. henselae strain Houston-1. Exploring these aspects is crucial for targeted therapeutic strategies against this versatile pathogen. Using reverse-vaccinology-based subtractive proteomics, this research aimed to identify the most antigenic proteins for formulating a multi-epitope vaccine against the B. henselae strain Houston-1. One crucial virulent and antigenic protein, the PAS domain-containing sensor histidine kinase protein, was identified. Subsequently, the identification of B-cell and T-cell epitopes for the specified protein was carried out and the evaluated epitopes were checked for their antigenicity, allergenicity, solubility, MHC binding capability, and toxicity. The filtered epitopes were merged using linkers and an adjuvant to create a multi-epitope vaccine construct. The structure was then refined, with 92.3% of amino acids falling within the allowed regions. Docking of the human receptor (TLR4) with the vaccine construct was performed and demonstrated a binding energy of -1047.2 Kcal/mol with more interactions. Molecular dynamic simulations confirmed the stability of this docked complex, emphasizing the conformation and interactions between the molecules. Further experimental validation is necessary to evaluate its effectiveness against B. henselae.
Collapse
Affiliation(s)
- Sudais Rahman
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan;
| | - Chien-Chun Chiou
- Department of Dermatology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan;
| | - Shabir Ahmad
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas (UNICAMP), Campinas 13084-862, Brazil;
| | - Zia Ul Islam
- Department of Biotechnology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan;
| |
Collapse
|
14
|
Ghiordanescu IM, Molinari N, Ciocănea-Teodorescu I, Schrijvers R, Motei C, Forsea AM, Demoly P, Chiriac AM. Skin Test Reactivity Patterns in Patients Allergic to Iodinated Contrast Media: A Refined View. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:705-713.e6. [PMID: 39056227 DOI: 10.1016/j.jaip.2023.10.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/30/2023] [Accepted: 10/23/2023] [Indexed: 07/28/2024]
Abstract
BACKGROUND Two-dimensional (2D) classifications of iodinated contrast media (ICM) are insufficient to explain the observed skin test (ST) reactivity patterns in patients with drug hypersensitivity reactions (DHRs) to ICM. OBJECTIVE To refine the current view on allergic DHRs to ICM by analyzing ST reactivity patterns in patients with previous reactions to ICM. METHODS Patients with a history of DHR to ICM and positive STs, who presented at the University Hospital of Montpellier between 2004 and 2022, were included in the study. The relative difference between every two ICM products was measured by Manhattan distance and odds ratios were computed for all pairs of products in the immediate reaction (IR) and non-immediate reaction (NIR) ST groups. RESULTS A total of 181 patients were included in the study. Odds ratio analysis identified significant associations between classical cross-reactive ICM, such as iohexol-ioversol, iohexol-iomeprol, iomeprol-ioversol, and iohexol-iodixanol in the IR ST group and iohexol-ioversol, iopromide-iohexol, and iomeprol-ioversol in the NIR ST group. We also identified uncommon associations, such as ioxitalamate-amidotrizoate in the IR ST group and amidotrizoate-iopamidol and amidotrizoate-ioxitalamate in the NIR ST group. The results were reflected by the Manhattan distance, which suggested the existence of clusters containing the same classically associated ICM as well as uncommon associations, which we hypothesize to be related to similarities in the 3D structure of the respective ICM. CONCLUSIONS Current chemical (2D) classifications cannot explain all observed ST reactivity patterns. Whether the 3D structure can be integrated into the current classifications to interpret the observed ST reactivity patterns and predict tolerance to alternative ICM requires further research.
Collapse
Affiliation(s)
- Ileana-Maria Ghiordanescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Institut Desbrest d'Épidémiologie et de Santé Publique, UMR UA11, University of Montpellier-INSERM, Montpellier, France; Dermatology and Allergy Clinic, Elias University Emergency Hospital, Bucharest, Romania.
| | - Nicolas Molinari
- Institut Desbrest d'Épidémiologie et de Santé Publique, UMR UA11, University of Montpellier-INSERM, Montpellier, France; Division of Medical Information, La Colombiere University Hospital of Montpellier, Montpellier, France
| | - Iuliana Ciocănea-Teodorescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Victor Babeș National Institute of Pathology, Bucharest, Romania
| | - Rik Schrijvers
- Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Cezara Motei
- Department of Pulmonology, Division of Allergy, Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France
| | - Ana-Maria Forsea
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Dermatology and Allergy Clinic, Elias University Emergency Hospital, Bucharest, Romania
| | - Pascal Demoly
- Institut Desbrest d'Épidémiologie et de Santé Publique, UMR UA11, University of Montpellier-INSERM, Montpellier, France; Department of Pulmonology, Division of Allergy, Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France
| | - Anca Mirela Chiriac
- Institut Desbrest d'Épidémiologie et de Santé Publique, UMR UA11, University of Montpellier-INSERM, Montpellier, France; Department of Pulmonology, Division of Allergy, Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France
| |
Collapse
|
15
|
Sives S, Keep S, Bickerton E, Vervelde L. Revealing Novel-Strain-Specific and Shared Epitopes of Infectious Bronchitis Virus Spike Glycoprotein Using Chemical Linkage of Peptides onto Scaffolds Precision Epitope Mapping. Viruses 2023; 15:2279. [PMID: 38005955 PMCID: PMC10675791 DOI: 10.3390/v15112279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
The avian coronavirus, infectious bronchitis virus (IBV), is an economically important infectious disease affecting chickens, with a diverse range of serotypes found globally. The major surface protein, spike (S), has high diversity between serotypes, and amino acid differences in the S1 sub-unit are thought to be responsible for poor cross-protection afforded by vaccination. Here, we attempt to address this, by using epitope mapping technology to identify shared and serotype-specific immunogenic epitopes of the S glycoprotein of three major circulating strains of IBV, M41, QX, and 4/91, via CLIPS peptide arrays based on peptides from the S1 sub-units. The arrays were screened with sera from chickens immunised with recombinant IBV, based on Beau-R backbone expressing heterologous S, generated in two independent vaccination/challenge trials. The screening of sera from rIBV vaccination experiments led to the identification of 52 immunogenic epitopes on the S1 of M41, QX, and 4/91. The epitopes were assigned into six overlapping epitope binding regions. Based on accessibility and location in the hypervariable regions of S, three sequences, 25YVYYYQSAFRPPNGWHLQGGAYAVVNSTN54, 67TVGVIKDVYNQSVASI82, and 83AMTVPPAGMSWSVS96, were selected for further investigation, and synthetic peptide mimics were recognised by polyclonal sera. These epitopes may have the potential to contribute towards a broader cross-protective IBV vaccine.
Collapse
Affiliation(s)
- Samantha Sives
- Division of Immunology, The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK
| | - Sarah Keep
- The Pirbright Institute, Ash Road, Woking GU24 0NF, UK
| | | | - Lonneke Vervelde
- Division of Immunology, The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK
| |
Collapse
|
16
|
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
|
17
|
Good MF, Yanow SK. Hiding in plain sight: an epitope-based strategy for a subunit malaria vaccine. Trends Parasitol 2023; 39:929-935. [PMID: 37684152 PMCID: PMC10592166 DOI: 10.1016/j.pt.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 09/10/2023]
Abstract
Recent data suggest that approaches to developing a subunit blood-stage malaria vaccine may be misdirected. While antigenic polymorphism is recognized as a challenge, efforts to counter this have primarily involved enhancing the quantity and quality of antibody with potent adjuvants, identifying conserved target proteins, or combining multiple antigens to broaden the immune response. However, paradoxically, evidence has emerged that narrowing, rather than broadening, the immune response may be required to obtain an immune response protective against multiple Plasmodium strains. Non-immunodominant, conserved epitopes are crucial. The evidence comes from studying the immune response to red cell surface-expressed antigens but should also be applicable to merozoite surface antigens. Strategies to define the targets of these highly focused immune responses are provided.
Collapse
Affiliation(s)
- Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast, Australia.
| | | |
Collapse
|
18
|
Hu D, Irving AT. Massively-multiplexed epitope mapping techniques for viral antigen discovery. Front Immunol 2023; 14:1192385. [PMID: 37818363 PMCID: PMC10561112 DOI: 10.3389/fimmu.2023.1192385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
Following viral infection, viral antigens bind specifically to receptors on the surface of lymphocytes thereby activating adaptive immunity in the host. An epitope, the smallest structural and functional unit of an antigen, binds specifically to an antibody or antigen receptor, to serve as key sites for the activation of adaptive immunity. The complexity and diverse range of epitopes are essential to study and map for the diagnosis of disease, the design of vaccines and for immunotherapy. Mapping the location of these specific epitopes has become a hot topic in immunology and immune therapy. Recently, epitope mapping techniques have evolved to become multiplexed, with the advent of high-throughput sequencing and techniques such as bacteriophage-display libraries and deep mutational scanning. Here, we briefly introduce the principles, advantages, and disadvantages of the latest epitope mapping techniques with examples for viral antigen discovery.
Collapse
Affiliation(s)
- Diya Hu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Aaron T. Irving
- Department of Clinical Laboratory Studies, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Centre for Infection, Immunity & Cancer, Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
- Biomedical and Health Translational Research Centre of Zhejiang Province (BIMET), Haining, China
- College of Medicine & Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
19
|
Zeng X, Bai G, Sun C, Ma B. Recent Progress in Antibody Epitope Prediction. Antibodies (Basel) 2023; 12:52. [PMID: 37606436 PMCID: PMC10443277 DOI: 10.3390/antib12030052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023] Open
Abstract
Recent progress in epitope prediction has shown promising results in the development of vaccines and therapeutics against various diseases. However, the overall accuracy and success rate need to be improved greatly to gain practical application significance, especially conformational epitope prediction. In this review, we examined the general features of antibody-antigen recognition, highlighting the conformation selection mechanism in flexible antibody-antigen binding. We recently highlighted the success and warning signs of antibody epitope predictions, including linear and conformation epitope predictions. While deep learning-based models gradually outperform traditional feature-based machine learning, sequence and structure features still provide insight into antibody-antigen recognition problems.
Collapse
Affiliation(s)
- Xincheng Zeng
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.Z.); (C.S.)
| | - Ganggang Bai
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.Z.); (C.S.)
| | - Chuance Sun
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.Z.); (C.S.)
| | - Buyong Ma
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.Z.); (C.S.)
- Shanghai Digiwiser Biological, Inc., Shanghai 200131, China
| |
Collapse
|
20
|
Karle AC, Wrobel MB, Koepke S, Gutknecht M, Gottlieb S, Christen B, Rubic-Schneider T, Pruimboom-Brees I, Leber XC, Scharenberg M, Maciejewski B, Turner O, Saravanan C, Huet F, Littlewood-Evans A, Clemens A, Grosskreutz CL, Kearns JD, Mehan P, Schmouder RL, Sasseville V, Brees D. Anti-brolucizumab immune response as one prerequisite for rare retinal vasculitis/retinal vascular occlusion adverse events. Sci Transl Med 2023; 15:eabq5241. [PMID: 36724238 DOI: 10.1126/scitranslmed.abq5241] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In October 2019, Novartis launched brolucizumab, a single-chain variable fragment molecule targeting vascular endothelial growth factor A, for the treatment of neovascular age-related macular degeneration. In 2020, rare cases of retinal vasculitis and/or retinal vascular occlusion (RV/RO) were reported, often during the first few months after treatment initiation, consistent with a possible immunologic pathobiology. This finding was inconsistent with preclinical studies in cynomolgus monkeys that demonstrated no drug-related intraocular inflammation, or RV/RO, despite the presence of preexisting and treatment-emergent antidrug antibodies (ADAs) in some animals. In this study, the immune response against brolucizumab in humans was assessed using samples from clinical trials and clinical practice. In the brolucizumab-naïve population, anti-brolucizumab ADA responses were detected before any treatment, which was supported by the finding that healthy donors can harbor brolucizumab-specific B cells. This suggested prior exposure of the immune system to proteins with structural similarity. Experiments on samples showed that naïve and brolucizumab-treated ADA-positive patients developed a class-switched, high-affinity immune response, with several linear epitopes being recognized by ADAs. Only patients with RV/RO showed a meaningful T cell response upon recall with brolucizumab. Further studies in cynomolgus monkeys preimmunized against brolucizumab with adjuvant followed by intravitreal brolucizumab challenge demonstrated that high ADA titers were required to generate ocular inflammation and vasculitis/vascular thrombosis, comparable to RV/RO in humans. Immunogenicity therefore seems to be a prerequisite to develop RV/RO. However, because only 2.1% of patients with ADA develop RV/RO, additional factors must play a role in the development of RV/RO.
Collapse
Affiliation(s)
- Anette C Karle
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Matthias B Wrobel
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Stephan Koepke
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Michael Gutknecht
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Sascha Gottlieb
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Brigitte Christen
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | | | | | | | - Meike Scharenberg
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | | | - Oliver Turner
- Novartis Institutes for BioMedical Research, East Hanover, NJ 07960, USA
| | - Chandra Saravanan
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Francois Huet
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | | | - Andreas Clemens
- Medical Affairs Region Europe, Novartis Pharma AG, Basel CH-4056, Switzerland
| | | | - Jeffrey D Kearns
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Pawan Mehan
- TRD Biologics & CGT, Novartis Pharma AG, Basel CH-4056, Switzerland
| | - Robert L Schmouder
- Novartis Institutes for BioMedical Research, East Hanover, NJ 07960, USA
| | - Vito Sasseville
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Dominique Brees
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| |
Collapse
|
21
|
Tsoumpeli MT, Varghese A, Owen JP, Maddison BC, Daly JM, Gough KC. Mapping Polyclonal Antibody Responses to Infection Using Next-Generation Phage Display. Methods Mol Biol 2023; 2702:467-487. [PMID: 37679636 DOI: 10.1007/978-1-0716-3381-6_25] [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: 09/09/2023]
Abstract
Peptide phage display has historically been used to epitope map monoclonal antibodies. More recently, by coupling this method with next-generation sequencing (so-called next-generation phage display, NGPD) to mass screen peptide binding events, the methodology has been successfully applied to map polyclonal antibody responses to infection. This leads to the identification of panels of mimotopes that represent the pathogen's epitopes. One potential advantage of using such an approach is that the mimotopes can represent not just linear epitopes but also conformational epitopes or those produced from post-translational modifications of proteins or from other non-protein macromolecules. The mapping of such complex immunological recognition of a pathogen can inform novel serological assay development and vaccine design. Here, we provide detailed methods for the application of NGPD to identify panels of mimotopes that are recognized specifically by antibodies from individuals with a particular infection.
Collapse
Affiliation(s)
- Maria T Tsoumpeli
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | - Anitha Varghese
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | | | | | - Janet M Daly
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | - Kevin C Gough
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, UK.
| |
Collapse
|
22
|
Lopes-Ribeiro Á, Araujo FP, Oliveira PDM, Teixeira LDA, Ferreira GM, Lourenço AA, Dias LCC, Teixeira CW, Retes HM, Lopes ÉN, Versiani AF, Barbosa-Stancioli EF, da Fonseca FG, Martins-Filho OA, Tsuji M, Peruhype-Magalhães V, Coelho-dos-Reis JGA. In silico and in vitro arboviral MHC class I-restricted-epitope signatures reveal immunodominance and poor overlapping patterns. Front Immunol 2022; 13:1035515. [PMID: 36466864 PMCID: PMC9713826 DOI: 10.3389/fimmu.2022.1035515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/31/2022] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION The present work sought to identify MHC-I-restricted peptide signatures for arbovirus using in silico and in vitro peptide microarray tools. METHODS First, an in-silico analysis of immunogenic epitopes restricted to four of the most prevalent human MHC class-I was performed by identification of MHC affinity score. For that, more than 10,000 peptide sequences from 5 Arbovirus and 8 different viral serotypes, namely Zika (ZIKV), Dengue (DENV serotypes 1-4), Chikungunya (CHIKV), Mayaro (MAYV) and Oropouche (OROV) viruses, in addition to YFV were analyzed. Haplotype HLA-A*02.01 was the dominant human MHC for all arboviruses. Over one thousand HLA-A2 immunogenic peptides were employed to build a comprehensive identity matrix. Intending to assess HLAA*02:01 reactivity of peptides in vitro, a peptide microarray was designed and generated using a dimeric protein containing HLA-A*02:01. RESULTS The comprehensive identity matrix allowed the identification of only three overlapping peptides between two or more flavivirus sequences, suggesting poor overlapping of virus-specific immunogenic peptides amongst arborviruses. Global analysis of the fluorescence intensity for peptide-HLA-A*02:01 binding indicated a dose-dependent effect in the array. Considering all assessed arboviruses, the number of DENV-derived peptides with HLA-A*02:01 reactivity was the highest. Furthermore, a lower number of YFV-17DD overlapping peptides presented reactivity when compared to non-overlapping peptides. In addition, the assessment of HLA-A*02:01-reactive peptides across virus polyproteins highlighted non-structural proteins as "hot-spots". Data analysis supported these findings showing the presence of major hydrophobic sites in the final segment of non-structural protein 1 throughout 2a (Ns2a) and in nonstructural proteins 2b (Ns2b), 4a (Ns4a) and 4b (Ns4b). DISCUSSION To our knowledge, these results provide the most comprehensive and detailed snapshot of the immunodominant peptide signature for arbovirus with MHC-class I restriction, which may bring insight into the design of future virus-specific vaccines to arboviruses and for vaccination protocols in highly endemic areas.
Collapse
Affiliation(s)
- Ágata Lopes-Ribeiro
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Franklin Pereira Araujo
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Patrícia de Melo Oliveira
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lorena de Almeida Teixeira
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovane Marques Ferreira
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alice Aparecida Lourenço
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laura Cardoso Corrêa Dias
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Caio Wilker Teixeira
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Henrique Morais Retes
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Élisson Nogueira Lopes
- Laboratorio de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Genética, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alice Freitas Versiani
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Pathology da University of Texas Medical Branch, Galveston, TX, United States
| | - Edel Figueiredo Barbosa-Stancioli
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Irving Medical School, Columbia University, New York City, NY, United States
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Jordana Grazziela Alves Coelho-dos-Reis
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| |
Collapse
|
23
|
Huan F, Gao S, Han TJ, Liu M, Li MS, Yang Y, Chen YY, Lai D, Cao MJ, Liu GM. Identification of the Immunoglobulin E Epitope of Arginine Kinase, an Important Allergen from Crassostrea angulata. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13419-13430. [PMID: 36205062 DOI: 10.1021/acs.jafc.2c05420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Arginine kinase (AK) was identified as an allergen in Crassostrea angulata. However, little information is available about its epitopes. In this study, AK from C. angulata was registered to the World Health Organization/International Union of Immunological Societies allergen nomenclature committee to be named as Cra a 2. The immunoglobulin G/immunoglobulin E-binding capacity of Cra a 2 was significantly reduced after chemical denaturation treatment. Further, eight linear mimotopes and five conformational mimotopes of Cra a 2 were obtained using phage panning. In addition to six linear epitopes that have been identified, two linear epitopes were verified by a synthetic peptide, of which L-Cra a 2-2 was conserved in shellfish. Four conformational epitopes were verified by site-directed mutation, among which mutation of C-Cra a 2-1 affected the structure and reduced the immunoreactivity of Cra a 2 most significantly. Overall, the identified epitopes may lay a foundation for the development of hypoallergenic oyster products through food processing.
Collapse
Affiliation(s)
- Fei Huan
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Shuai Gao
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Tian-Jiao Han
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Meng Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Meng-Si Li
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Yang Yang
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Yi-Yu Chen
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Dong Lai
- Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian 361021, People's Republic of China
| | - Min-Jie Cao
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Guang-Ming Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| |
Collapse
|
24
|
Zhou J, Chen J, Peng Y, Xie Y, Xiao Y. A Promising Tool in Serological Diagnosis: Current Research Progress of Antigenic Epitopes in Infectious Diseases. Pathogens 2022; 11:1095. [PMID: 36297152 PMCID: PMC9609281 DOI: 10.3390/pathogens11101095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 07/30/2023] Open
Abstract
Infectious diseases, caused by various pathogens in the clinic, threaten the safety of human life, are harmful to physical and mental health, and also increase economic burdens on society. Infections are a complex mechanism of interaction between pathogenic microorganisms and their host. Identification of the causative agent of the infection is vital for the diagnosis and treatment of diseases. Etiological laboratory diagnostic tests are therefore essential to identify pathogens. However, due to its rapidity and automation, the serological diagnostic test is among the methods of great significance for the diagnosis of infections with the basis of detecting antigens or antibodies in body fluids clinically. Epitopes, as a special chemical group that determines the specificity of antigens and the basic unit of inducing immune responses, play an important role in the study of immune responses. Identifying the epitopes of a pathogen may contribute to the development of a vaccine to prevent disease, the diagnosis of the corresponding disease, and the determination of different stages of the disease. Moreover, both the preparation of neutralizing antibodies based on useful epitopes and the assembly of several associated epitopes can be used in the treatment of disease. Epitopes can be divided into B cell epitopes and T cell epitopes; B cell epitopes stimulate the body to produce antibodies and are therefore commonly used as targets for the design of serological diagnostic experiments. Meanwhile, epitopes can fall into two possible categories: linear and conformational. This article reviews the role of B cell epitopes in the clinical diagnosis of infectious diseases.
Collapse
|
25
|
Lemes MR, Rodrigues TCV, Jaiswal AK, Tiwari S, Sales-Campos H, Andrade-Silva LE, Oliveira CJF, Azevedo V, Rodrigues V, Soares SC, da Silva MV. In silico designing of a recombinant multi-epitope antigen for leprosy diagnosis. J Genet Eng Biotechnol 2022; 20:128. [PMID: 36053342 PMCID: PMC9440174 DOI: 10.1186/s43141-022-00411-7] [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: 08/24/2021] [Accepted: 08/25/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Leprosy is caused by Mycobacterium leprae and Mycobacterium lepromatosis. Most of the affected population lives in low-income countries and may take up to 10 years to show any clinical signs, which is how physicians diagnose it. However, due to progressive cell damage, early diagnosis is very important. The best way to confirm leprosy is through bacilloscopic, which only confirms the diagnosis and has low accuracy or PCR, that requires specialized operators and is expensive. Since the bacteria are fastidious and do not grow in any culture media, therefore, diagnosing leprosy in the lab is still a challenge. In this concern, a recombinant multi-epitope protein can be a beneficial strategy in the management of the diagnosis, as diverse immunogenic epitopes are precisely selected to detect specific antibodies. Therefore, the purposes of the present study were to select immunogenic epitopes from different relevant proteins, with immunogenic properties, and then to construct a recombinant multi-epitope protein that accuses the presence of the antibodies in the early stages of the disease, making it more than appropriate to be applied as a diagnostic tool. RESULTS We selected 22 common proteins from both species and, using bioinformatics tools, predicted B and T cell epitopes. After multiple filtering and analyzing, we ended up with 29 epitopes {MHC-I (total 18) and MHC-II (total 11)} from 10 proteins, which were then merged into one construct. Its secondary and tertiary structures were also predicted and refined to comprise the amino acid residues in the best conformation possible. The multi-epitope protein construct was stable, non-host homologous, non-allergic, non-toxic, and elicit humoral and cellular responses. It has conformational B cell epitopes and potential to elicit IFN-γ, IL-4, and IL-10 secretion. CONCLUSIONS This novel recombinant multi-epitope protein constructed using the common epitopes from M. leprae and M. lepromatosis has a huge immunological potential, is stable, and can be lyophilized to be used in ELISA plates or even in biosensors, which are user-friendly diagnosis tools, facilitating translation into human sample tests.
Collapse
Affiliation(s)
- Marcela Rezende Lemes
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-180, Brazil
| | - Thaís Cristina Vilela Rodrigues
- Laboratory of Cellular and Molecular Genetics (LGCM) Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences,, Federal University of Minas Gerais (UFMG), MG, 31270-901, Belo Horizonte, Brazil
| | - Arun Kumar Jaiswal
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-180, Brazil
- Laboratory of Cellular and Molecular Genetics (LGCM) Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences,, Federal University of Minas Gerais (UFMG), MG, 31270-901, Belo Horizonte, Brazil
| | - Sandeep Tiwari
- Laboratory of Cellular and Molecular Genetics (LGCM) Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences,, Federal University of Minas Gerais (UFMG), MG, 31270-901, Belo Horizonte, Brazil.
| | - Helioswilton Sales-Campos
- Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Goiás, Brazil
| | - Leonardo Eurípedes Andrade-Silva
- Infectious Disease Department, Institute of Health Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Carlo Jose Freire Oliveira
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-180, Brazil
| | - Vasco Azevedo
- Laboratory of Cellular and Molecular Genetics (LGCM) Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences,, Federal University of Minas Gerais (UFMG), MG, 31270-901, Belo Horizonte, Brazil
| | - Virmondes Rodrigues
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-180, Brazil
| | - Siomar C Soares
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-180, Brazil
| | - Marcos Vinicius da Silva
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-180, Brazil.
| |
Collapse
|
26
|
Kang W, Zhang J, Li H, Yu N, Tang R, Sun X, He L, Sun J, Chen Y. Identification of Major B-Cell Linear Epitopes of Peach Allergen Pru p 3 Using Immune Slot-Blot Microarray Assay. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8134-8144. [PMID: 35749217 DOI: 10.1021/acs.jafc.2c01448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pru p 3, one of the most representative proteins of the lipid transfer proteins (LTPs), is responsible for clinical allergic reactions to food of peach origin. The identification of Pru p 3 epitopes is not comprehensive due to different methods and principles of epitope screening. In addition, evaluation of the stability of the epitopes and the validation of the immunological key amino acids still need further research. Therefore, in the present study, an immune slot-blot microarray assay was performed to screen the epitopes from Pru p 3 overlapping peptide library, and a new epitope (P-1, AA1-16, ITCGQVSSALAPCIPY) was identified and two identified epitopes were deeply investigated (P-2, AA12-27, PCIPYVRGGGAVPPAC; P-3, AA23-38, VPPACCNGIRNVNNLA). The stability of these epitopes was then verified by thermal processing treatment and digestion experiments. Moreover, the key amino acids of the three identified epitopes were obtained by epitope amino acid mutation combined with slot-blot experiments. These findings may contribute to the further understanding of Pru p 3 and the prevention of peach allergy.
Collapse
Affiliation(s)
- Wenhan Kang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, School of Food Science Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi 214122, People's Republic of China
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Jiukai Zhang
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Hong Li
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, People's Republic of China
| | - Ning Yu
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Rui Tang
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, People's Republic of China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, School of Food Science Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi 214122, People's Republic of China
| | - Lei He
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| | - Jinlyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, People's Republic of China
| | - Ying Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, School of Food Science Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi 214122, People's Republic of China
- Chinese Academy of Inspection and Quarantine, Beijing 100176, People's Republic of China
| |
Collapse
|
27
|
de Figueiredo DB, Kaneko K, Rodrigues TDC, MacLoughlin R, Miyaji EN, Saleem I, Gonçalves VM. Pneumococcal Surface Protein A-Hybrid Nanoparticles Protect Mice from Lethal Challenge after Mucosal Immunization Targeting the Lungs. Pharmaceutics 2022; 14:pharmaceutics14061238. [PMID: 35745810 PMCID: PMC9230107 DOI: 10.3390/pharmaceutics14061238] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 12/04/2022] Open
Abstract
Pneumococcal disease remains a global burden, with current conjugated vaccines offering protection against the common serotype strains. However, there are over 100 serotype strains, and serotype replacement is now being observed, which reduces the effectiveness of the current vaccines. Pneumococcal surface protein A (PspA) has been investigated as a candidate for new serotype-independent pneumococcal vaccines, but requires adjuvants and/or delivery systems to improve protection. Polymeric nanoparticles (NPs) are biocompatible and, besides the antigen, can incorporate mucoadhesive and adjuvant substances such as chitosans, which improve antigen presentation at mucosal surfaces. This work aimed to define the optimal NP formulation to deliver PspA into the lungs and protect mice against lethal challenge. We prepared poly(glycerol-adipate-co-ω-pentadecalactone) (PGA-co-PDL) and poly(lactic-co-glycolic acid) (PLGA) NPs using an emulsion/solvent evaporation method, incorporating chitosan hydrochloride (HCl-CS) or carboxymethyl chitosan (CM-CS) as hybrid NPs with encapsulated or adsorbed PspA. We investigated the physicochemical properties of NPs, together with the PspA integrity and biological activity. Furthermore, their ability to activate dendritic cells in vitro was evaluated, followed by mucosal immunization targeting mouse lungs. PGA-co-PDL/HCl-CS (291 nm) or CM-CS (281 nm) NPs produced smaller sizes compared to PLGA/HCl-CS (310 nm) or CM-CS (299 nm) NPs. Moreover, NPs formulated with HCl-CS possessed a positive charge (PGA-co-PDL +17 mV, PLGA + 13 mV) compared to those formulated with CM-CS (PGA-co-PDL -20 mV, PLGA -40 mV). PspA released from NPs formulated with HCl-CS preserved the integrity and biological activity, but CM-CS affected PspA binding to lactoferrin and antibody recognition. PspA adsorbed in PGA-co-PDL/HCl-CS NPs stimulated CD80+ and CD86+ cells, but this was lower compared to when PspA was encapsulated in PLGA/HCl-CS NPs, which also stimulated CD40+ and MHC II (I-A/I-E)+ cells. Despite no differences in IgG being observed between immunized animals, PGA-co-PDL/HCl-CS/adsorbed-PspA protected 83% of mice after lethal pneumococcal challenge, while 100% of mice immunized with PLGA/HCl-CS/encapsulated-PspA were protected. Therefore, this formulation is a promising vaccine strategy, which has beneficial properties for mucosal immunization and could potentially provide serotype-independent protection.
Collapse
Affiliation(s)
- Douglas Borges de Figueiredo
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-070, Brazil;
| | - Kan Kaneko
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK;
| | - Tasson da Costa Rodrigues
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-070, Brazil;
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen, IDA Business Park, H91 HE94 Galway, Ireland;
| | - Eliane Namie Miyaji
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Imran Saleem
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK;
- Correspondence: (I.S.); (V.M.G.); Tel.: +55-112-6279819 (V.M.G.)
| | - Viviane Maimoni Gonçalves
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Correspondence: (I.S.); (V.M.G.); Tel.: +55-112-6279819 (V.M.G.)
| |
Collapse
|
28
|
Identification of Oocyst-Driven Toxoplasma gondii Infections in Humans and Animals through Stage-Specific Serology-Current Status and Future Perspectives. Microorganisms 2021; 9:microorganisms9112346. [PMID: 34835471 PMCID: PMC8618849 DOI: 10.3390/microorganisms9112346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
The apicomplexan zoonotic parasite Toxoplasma gondii has three infective stages: sporozoites in sporulated oocysts, which are shed in unsporulated form into the environment by infected felids; tissue cysts containing bradyzoites, and fast replicating tachyzoites that are responsible for acute toxoplasmosis. The contribution of oocysts to infections in both humans and animals is understudied despite being highly relevant. Only a few diagnostic antigens have been described to be capable of discriminating which parasite stage has caused an infection. Here we provide an extensive overview of the antigens and serological assays used to detect oocyst-driven infections in humans and animals according to the literature. In addition, we critically discuss the possibility to exploit the increasing knowledge of the T. gondii genome and the various 'omics datasets available, by applying predictive algorithms, for the identification of new oocyst-specific proteins for diagnostic purposes. Finally, we propose a workflow for how such antigens and assays based on them should be evaluated to ensure reproducible and robust results.
Collapse
|
29
|
Mariotti S, Capocefalo A, Chiantore MV, Iacobino A, Teloni R, De Angelis ML, Gallinaro A, Pirillo MF, Borghi M, Canitano A, Michelini Z, Baggieri M, Marchi A, Bucci P, McKay PF, Acchioni C, Sandini S, Sgarbanti M, Tosini F, Di Virgilio A, Venturi G, Marino F, Esposito V, Di Bonito P, Magurano F, Cara A, Negri D, Nisini R. Isolation and Characterization of Mouse Monoclonal Antibodies That Neutralize SARS-CoV-2 and Its Variants of Concern Alpha, Beta, Gamma and Delta by Binding Conformational Epitopes of Glycosylated RBD With High Potency. Front Immunol 2021; 12:750386. [PMID: 34764961 PMCID: PMC8576447 DOI: 10.3389/fimmu.2021.750386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 01/14/2023] Open
Abstract
Antibodies targeting Receptor Binding Domain (RBD) of SARS-CoV-2 have been suggested to account for the majority of neutralizing activity in COVID-19 convalescent sera and several neutralizing antibodies (nAbs) have been isolated, characterized and proposed as emergency therapeutics in the form of monoclonal antibodies (mAbs). However, SARS-CoV-2 variants are rapidly spreading worldwide from the sites of initial identification. The variants of concern (VOC) B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma) and B.1.167.2 (Delta) showed mutations in the SARS-CoV-2 spike protein potentially able to cause escape from nAb responses with a consequent reduction of efficacy of vaccines and mAbs-based therapy. We produced the recombinant RBD (rRBD) of SARS-CoV-2 spike glycoprotein from the Wuhan-Hu 1 reference sequence in a mammalian system, for mice immunization to isolate new mAbs with neutralizing activity. Here we describe four mAbs that were able to bind the rRBD in Enzyme-Linked Immunosorbent Assay and the transmembrane full-length spike protein expressed in HEK293T cells by flow cytometry assay. Moreover, the mAbs recognized the RBD in supernatants of SARS-CoV-2 infected VERO E6 cells by Western Blot under non-reducing condition or in supernatants of cells infected with lentivirus pseudotyped for spike protein, by immunoprecipitation assay. Three out of four mAbs lost their binding efficiency to completely N-deglycosylated rRBD and none was able to bind the same recombinant protein expressed in Escherichia coli, suggesting that the epitopes recognized by three mAbs are generated by the conformational structure of the glycosylated native protein. Of particular relevance, three mAbs were able to inhibit Wuhan SARS-CoV-2 infection of VERO E6 cells in a plaque-reduction neutralization test and the Wuhan SARS-CoV-2 as well as the Alpha, Beta, Gamma and Delta VOC in a pseudoviruses-based neutralization test. These mAbs represent important additional tools for diagnosis and therapy of COVID-19 and may contribute to the understanding of the functional structure of SARS-CoV-2 RBD.
Collapse
Affiliation(s)
- Sabrina Mariotti
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Antonio Capocefalo
- Dipartimento Sicurezza alimentare, nutrizione e sanità pubblica veterinaria, Istituto Superiore di Sanità, Roma, Italy
| | | | - Angelo Iacobino
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Raffaela Teloni
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Maria Laura De Angelis
- Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Roma, Italy
| | - Alessandra Gallinaro
- Centro nazionale per la salute globale, Istituto Superiore di Sanità, Roma, Italy
| | - Maria Franca Pirillo
- Centro nazionale per la salute globale, Istituto Superiore di Sanità, Roma, Italy
| | - Martina Borghi
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Andrea Canitano
- Centro nazionale per la salute globale, Istituto Superiore di Sanità, Roma, Italy
| | - Zuleika Michelini
- Centro nazionale per la salute globale, Istituto Superiore di Sanità, Roma, Italy
| | - Melissa Baggieri
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Antonella Marchi
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Paola Bucci
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Paul F. McKay
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Chiara Acchioni
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Silvia Sandini
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Marco Sgarbanti
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Fabio Tosini
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Antonio Di Virgilio
- Centro per la sperimentazione ed il benessere animale, Istituto Superiore di Sanità, Roma, Italy
| | - Giulietta Venturi
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Francesco Marino
- Centro nazionale per il controllo e la valutazione dei farmaci, Istituto Superiore di Sanità, Roma, Italy
| | - Valeria Esposito
- Centro nazionale per il controllo e la valutazione dei farmaci, Istituto Superiore di Sanità, Roma, Italy
| | - Paola Di Bonito
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Fabio Magurano
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Andrea Cara
- Centro nazionale per la salute globale, Istituto Superiore di Sanità, Roma, Italy
| | - Donatella Negri
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie infettive, Istituto Superiore di Sanità, Roma, Italy
| |
Collapse
|
30
|
Qiao X, Qu L, Guo Y, Hoshino T. Secondary Structure and Conformational Stability of the Antigen Residues Making Contact with Antibodies. J Phys Chem B 2021; 125:11374-11385. [PMID: 34615354 DOI: 10.1021/acs.jpcb.1c05997] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antibodies are crucial biomolecules that bring high therapeutic efficacy in medicine and accurate molecular detection in diagnosis. Many studies have been devoted to analyzing the antigen-antibody interaction from the importance of understanding the antibody recognition mechanism. However, most of the previous studies examined the characteristic of the antibody for interaction. It is also informative to clarify the significant antigen residues contributing to the binding. To characterize the molecular interaction of antigens, we computationally analyzed 350 antigen-antibody complex structures by molecular mechanics (MM) calculations and molecular dynamics (MD) simulations. Based on the MM calculations, the antigen residues contributing to the binding were extracted from all the 350 complexes. The extracted residues are located at the antigen-antibody interface and are responsible for making contact with the antibody. The appearances of the charged polar residues, Asp, Glu, Arg, and Lys, were noticeably large. In contrast, the populations of the hydrophobic residues, Leu, Val, and Ala, were relatively low. The appearance frequencies of the other amino acid residues were almost close to the abundance of general proteins of eukaryotes. The binding score indicated that the hydrophilic interaction was dominant at the antigen-antibody contact instead of the hydrophobic one. The positively charged residues, Arg and Lys, remarkably contributed to the binding compared to the negatively charged ones, Asp and Glu. Considerable contributions were also observed for the noncharged polar residues, Asn and Gln. The analysis of the secondary structures of the extracted antigen residues suggested that there was no marked difference in recognition by antibodies among helix, sheet, turn, and coil. A long helix of the antigen sometimes made contact with antibody complementarity-determining regions, and a large sheet also frequently covered the antibody heavy and light chains. The turn structure was the most popularly observed at the contact with antibody among 350 complexes. Three typical complexes were picked up for each of the four secondary structures. MD simulations were performed to examine the stability of the interfacial structures of the antigens for these 12 complex models. The alterations of secondary structures were monitored through the simulations. The structural fluctuations of the contact residues were low compared with the other domains of antigen molecules. No drastic conversion was observed for every model during the 100 ns simulation. The motions of the interfacial antigen residues were small compared to the other residues on the protein surface. Therefore, diverse molecular conformations are possible for antibody recognition as long as the target areas are polar, nonflexible, and protruding on the protein surface.
Collapse
Affiliation(s)
- Xinyue Qiao
- Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| | - Liang Qu
- Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| | - Yan Guo
- Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| | - Tyuji Hoshino
- Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
31
|
Rawal K, Sinha R, Abbasi BA, Chaudhary A, Nath SK, Kumari P, Preeti P, Saraf D, Singh S, Mishra K, Gupta P, Mishra A, Sharma T, Gupta S, Singh P, Sood S, Subramani P, Dubey AK, Strych U, Hotez PJ, Bottazzi ME. Identification of vaccine targets in pathogens and design of a vaccine using computational approaches. Sci Rep 2021; 11:17626. [PMID: 34475453 PMCID: PMC8413327 DOI: 10.1038/s41598-021-96863-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Antigen identification is an important step in the vaccine development process. Computational approaches including deep learning systems can play an important role in the identification of vaccine targets using genomic and proteomic information. Here, we present a new computational system to discover and analyse novel vaccine targets leading to the design of a multi-epitope subunit vaccine candidate. The system incorporates reverse vaccinology and immuno-informatics tools to screen genomic and proteomic datasets of several pathogens such as Trypanosoma cruzi, Plasmodium falciparum, and Vibrio cholerae to identify potential vaccine candidates (PVC). Further, as a case study, we performed a detailed analysis of the genomic and proteomic dataset of T. cruzi (CL Brenner and Y strain) to shortlist eight proteins as possible vaccine antigen candidates using properties such as secretory/surface-exposed nature, low transmembrane helix (< 2), essentiality, virulence, antigenic, and non-homology with host/gut flora proteins. Subsequently, highly antigenic and immunogenic MHC class I, MHC class II and B cell epitopes were extracted from top-ranking vaccine targets. The designed vaccine construct containing 24 epitopes, 3 adjuvants, and 4 linkers was analysed for its physicochemical properties using different tools, including docking analysis. Immunological simulation studies suggested significant levels of T-helper, T-cytotoxic cells, and IgG1 will be elicited upon administration of such a putative multi-epitope vaccine construct. The vaccine construct is predicted to be soluble, stable, non-allergenic, non-toxic, and to offer cross-protection against related Trypanosoma species and strains. Further, studies are required to validate safety and immunogenicity of the vaccine.
Collapse
Affiliation(s)
- Kamal Rawal
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India.
| | - Robin Sinha
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Bilal Ahmed Abbasi
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Amit Chaudhary
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Swarsat Kaushik Nath
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Priya Kumari
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - P Preeti
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Devansh Saraf
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Shachee Singh
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Kartik Mishra
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Pranjay Gupta
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Astha Mishra
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Trapti Sharma
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Srijanee Gupta
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Prashant Singh
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Shriya Sood
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Preeti Subramani
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Aman Kumar Dubey
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, USA
| |
Collapse
|
32
|
Sahlström P, Hansson M, Steen J, Amara K, Titcombe PJ, Forsström B, Stålesen R, Israelsson L, Piccoli L, Lundberg K, Klareskog L, Mueller DL, Catrina AI, Skriner K, Malmström V, Grönwall C. Different Hierarchies of Anti–Modified Protein Autoantibody Reactivities in Rheumatoid Arthritis. Arthritis Rheumatol 2020; 72:1643-1657. [DOI: 10.1002/art.41385] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 05/13/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Peter Sahlström
- Karolinska Institutet Karolinska University Hospital, Stockholm, Sweden, and Charité Universitätsmedizin Berlin Germany
| | - Monika Hansson
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Johanna Steen
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Khaled Amara
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Philip J. Titcombe
- Karolinska Institutet Karolinska University Hospital, Stockholm, Sweden, and University of Minnesota Medical School Minneapolis
| | | | - Ragnhild Stålesen
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Lena Israelsson
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Luca Piccoli
- Institute for Research in Biomedicine Università della Svizzera italiana Bellinzona Switzerland
| | - Karin Lundberg
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Lars Klareskog
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | | | - Anca I. Catrina
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | | | | | - Caroline Grönwall
- Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
33
|
Mikus M, Zandian A, Sjöberg R, Hamsten C, Forsström B, Andersson M, Greiff L, Uhlén M, Levin M, Nilsson P, van Hage M, Ohlin M. Allergome-wide peptide microarrays enable epitope deconvolution in allergen-specific immunotherapy. J Allergy Clin Immunol 2020; 147:1077-1086. [PMID: 32791163 DOI: 10.1016/j.jaci.2020.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 07/22/2020] [Accepted: 08/04/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The interaction of allergens and allergen-specific IgE initiates the allergic cascade after crosslinking of receptors on effector cells. Antibodies of other isotypes may modulate such a reaction. Receptor crosslinking requires binding of antibodies to multiple epitopes on the allergen. Limited information is available on the complexity of the epitope structure of most allergens. OBJECTIVES We sought to allow description of the complexity of IgE, IgG4, and IgG epitope recognition at a global, allergome-wide level during allergen-specific immunotherapy (AIT). METHODS We generated an allergome-wide microarray comprising 731 allergens in the form of more than 172,000 overlapping 16-mer peptides. Allergen recognition by IgE, IgG4, and IgG was examined in serum samples collected from subjects undergoing AIT against pollen allergy. RESULTS Extensive induction of linear peptide-specific Phl p 1- and Bet v 1-specific humoral immunity was demonstrated in subjects undergoing a 3-year-long AIT against grass and birch pollen allergy, respectively. Epitope profiles differed between subjects but were largely established already after 1 year of AIT, suggesting that dominant allergen-specific antibody clones remained as important contributors to humoral immunity following their initial establishment during the early phase of AIT. Complex, subject-specific patterns of allergen isoform and group cross-reactivities in the repertoires were observed, patterns that may indicate different levels of protection against different allergen sources. CONCLUSIONS The study highlights the complexity and subject-specific nature of allergen epitopes recognized following AIT. We envisage that epitope deconvolution will be an important aspect of future efforts to describe and analyze the outcomes of AIT in a personalized manner.
Collapse
Affiliation(s)
- Maria Mikus
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology & SciLifeLab, Stockholm, Sweden; Unit of Experimental Asthma and Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Arash Zandian
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology & SciLifeLab, Stockholm, Sweden
| | - Ronald Sjöberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology & SciLifeLab, Stockholm, Sweden
| | - Carl Hamsten
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Björn Forsström
- Division of Systems Biology, Department of Protein Science, KTH Royal Institute of Technology & SciLifeLab, Stockholm, Sweden
| | - Morgan Andersson
- Department of Otorhinolaryngology, Head & Neck Surgery, Skåne University Hospital, Lund, Sweden
| | - Lennart Greiff
- Department of Otorhinolaryngology, Head & Neck Surgery, Skåne University Hospital, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Mathias Uhlén
- Division of Systems Biology, Department of Protein Science, KTH Royal Institute of Technology & SciLifeLab, Stockholm, Sweden
| | - Mattias Levin
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology & SciLifeLab, Stockholm, Sweden
| | - Marianne van Hage
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Mats Ohlin
- Department of Immunotechnology, Lund University, Lund, Sweden.
| |
Collapse
|
34
|
Dong R, Chu Z, Yu F, Zha Y. Contriving Multi-Epitope Subunit of Vaccine for COVID-19: Immunoinformatics Approaches. Front Immunol 2020; 11:1784. [PMID: 32849643 PMCID: PMC7399176 DOI: 10.3389/fimmu.2020.01784] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 01/10/2023] Open
Abstract
COVID-19 has recently become the most serious threat to public health, and its prevalence has been increasing at an alarming rate. The incubation period for the virus is ~1-14 days and all age groups may be susceptible to a fatality rate of about 5.9%. COVID-19 is caused by a novel single-stranded, positive (+) sense RNA beta coronavirus. The development of a vaccine for SARS-CoV-2 is an urgent need worldwide. Immunoinformatics approaches are both cost-effective and convenient, as in silico predictions can reduce the number of experiments needed. In this study, with the aid of immunoinformatics tools, we tried to design a multi-epitope vaccine that can be used for the prevention and treatment of COVID-19. The epitopes were computed by using B cells, cytotoxic T lymphocytes (CTL), and helper T lymphocytes (HTL) base on the proteins of SARS-CoV-2. A vaccine was devised by fusing together the B cell, HTL, and CTL epitopes with linkers. To enhance the immunogenicity, the β-defensin (45 mer) amino acid sequence, and pan-HLA DR binding epitopes (13aa) were adjoined to the N-terminal of the vaccine with the help of the EAAAK linker. To enable the intracellular delivery of the modeled vaccine, a TAT sequence (11aa) was appended to C-terminal. Linkers play vital roles in producing an extended conformation (flexibility), protein folding, and separation of functional domains, and therefore, make the protein structure more stable. The secondary and three-dimensional (3D) structure of the final vaccine was then predicted. Furthermore, the complex between the final vaccine and immune receptors (toll-like receptor-3 (TLR-3), major histocompatibility complex (MHC-I), and MHC-II) were evaluated by molecular docking. Lastly, to confirm the expression of the designed vaccine, the mRNA of the vaccine was enhanced with the aid of the Java Codon Adaptation Tool, and the secondary structure was generated from Mfold. Then we performed in silico cloning. The final vaccine requires experimental validation to determine its safety and efficacy in controlling SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Rong Dong
- Department of Biomedicine, Guizhou University School of Medicine, Guiyang, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases (Guizhou Provincial People's Hospital), Guiyang, China
| | - Zhugang Chu
- Department of Urinary Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Fuxun Yu
- NHC Key Laboratory of Pulmonary Immunological Diseases (Guizhou Provincial People's Hospital), Guiyang, China
| | - Yan Zha
- Department of Biomedicine, Guizhou University School of Medicine, Guiyang, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases (Guizhou Provincial People's Hospital), Guiyang, China
| |
Collapse
|