1
|
Zúñiga E, Contreras-Trigo B, Buchert J, Sáez-Ahumada F, Hernández L, Fica-León V, Nova-Lamperti E, Kobe B, Guzmán F, Diaz-García V, Guzmán-Gutiérrez E, Oyarzún P. Gold Nanoparticles as a Platform for Delivery of Immunogenic Peptides to THP-1 Derived Macrophages: Insights into Nanotoxicity. Vaccines (Basel) 2025; 13:119. [PMID: 40006666 PMCID: PMC11860437 DOI: 10.3390/vaccines13020119] [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: 12/14/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Peptide-based nanovaccines have emerged as a promising strategy for combating infectious diseases, as they overcome the low immunogenicity that is inherent to short epitope-containing synthetic peptides. Gold nanoparticles (AuNPs) present several advantages as peptide nanocarriers, but a deeper understanding of the design criteria is paramount to accelerate the development of peptide-AuNPs nanoconjugates (p-AuNPs). METHODS Herein, we synthesized and characterized p-AuNPs of 23 nm (p-Au23) and 68 nm (p-Au68) with varying levels of peptide surface coverage and different peptide designs, investigating their effect on the cell viability (cell death and mitochondrial activity), cellular uptake, and cathepsin B activity in THP-1 macrophages. RESULTS p-Au23 proved no negative effect in the cell viability and high levels of nanoconjugate uptake, but p-Au68 induced strong toxicity to the cell line. The peptide sequences were successfully designed with spacer regions and a cell-penetrating peptide (pTAT) that enhanced cellular uptake and cathepsin B activity for p-Au23, while pTAT induced severe effects in the THP-1 viability (~40-60% cell death). CONCLUSIONS These findings provide valuable insight into the design criteria of AuNPs and immunogenic peptides, along with nanotoxicity effects associated with AuNP size and surface charge in human monocyte-derived macrophages.
Collapse
Affiliation(s)
- Eduardo Zúñiga
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Braulio Contreras-Trigo
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Jorge Buchert
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Fabián Sáez-Ahumada
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Leonardo Hernández
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Víctor Fica-León
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Estefania Nova-Lamperti
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Fanny Guzmán
- Núcleo Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile;
| | - Víctor Diaz-García
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| | - Enrique Guzmán-Gutiérrez
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile; (J.B.); (F.S.-A.); (L.H.); (E.N.-L.); (E.G.-G.)
| | - Patricio Oyarzún
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción 4081339, Chile; (E.Z.); (B.C.-T.); (V.F.-L.); (V.D.-G.)
| |
Collapse
|
2
|
Öztemiz Topcu E, Gadermaier G. To stay or not to stay intact as an allergen: the endolysosomal degradation assay used as tool to analyze protein immunogenicity and T cell epitopes. FRONTIERS IN ALLERGY 2024; 5:1440360. [PMID: 39071040 PMCID: PMC11272489 DOI: 10.3389/falgy.2024.1440360] [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: 05/29/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Antigen uptake and processing of exogenous proteins is critical for adaptive immunity, particularly for T helper cell activation. Proteins undergo distinct proteolytic processing in endolysosomal compartments of antigen-presenting cells. The resulting peptides are presented on MHC class II molecules and specifically recognized by T cells. The in vitro endolysosomal degradation assay mimics antigen processing by incubating a protein of interest with a protease cocktail derived from the endolysosomal compartments of antigen presenting cells. The kinetics of protein degradation is monitored by gel electrophoresis and allows calculation of a protein's half-life and thus endolysosomal stability. Processed peptides are analyzed by mass spectrometry and abundant peptide clusters are shown to harbor T cell epitopes. The endolysosomal degradation assay has been widely used to study allergens, which are IgE-binding proteins involved in type I hypersensitivity. In this review article, we provide the first comprehensive overview of the endolysosomal degradation of 29 isoallergens and variants originating from the PR-10, Ole e 1-like, pectate lyase, defensin polyproline-linked, non-specific lipid transfer, mite group 1, 2, and 5, and tropomyosin protein families. The assay method is described in detail and suggestions for improved standardization and reproducibility are provided. The current hypothesis implies that proteins with high endolysosomal stability can induce an efficient immune response, whereas highly unstable proteins are degraded early during antigen processing and therefore not efficient for MHC II peptide presentation. To validate this concept, systematic analyses of high and low allergenic representatives of protein families should be investigated. In addition to purified molecules, allergen extracts should be degraded to analyze potential matrix effects and gastrointestinal proteolysis of food allergens. In conclusion, individual protein susceptibility and peptides obtained from the endolysosomal degradation assay are powerful tools for understanding protein immunogenicity and T cell reactivity. Systematic studies and linkage with in vivo sensitization data will allow the establishment of (machine-learning) tools to aid prediction of immunogenicity and allergenicity. The orthogonal method could in the future be used for risk assessment of novel foods and in the generation of protein-based immunotherapeutics.
Collapse
|
3
|
Halvorson T, Ivison S, Huang Q, Ladua G, Yotis DM, Mannar D, Subramaniam S, Ferreira VH, Kumar D, Belga S, Levings MK. SARS-CoV-2 Variants Omicron BA.4/5 and XBB.1.5 Significantly Escape T Cell Recognition in Solid-organ Transplant Recipients Vaccinated Against the Ancestral Strain. Transplantation 2024; 108:e49-e62. [PMID: 38012843 DOI: 10.1097/tp.0000000000004873] [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] [Indexed: 11/29/2023]
Abstract
BACKGROUND Immune-suppressed solid-organ transplant recipients (SOTRs) display impaired humoral responses to COVID-19 vaccination, but T cell responses are incompletely understood. SARS-CoV-2 variants Omicron BA.4/5 (BA.4/5) and XBB.1.5 escape neutralization by antibodies induced by vaccination or infection with earlier strains, but T cell recognition of these lineages in SOTRs is unclear. METHODS We characterized Spike-specific T cell responses to ancestral SARS-CoV-2 and BA.4/5 peptides in 42 kidney, liver, and lung transplant recipients throughout a 3- or 4-dose ancestral Spike mRNA vaccination schedule. As the XBB.1.5 variant emerged during the study, we tested vaccine-induced T cell responses in 10 additional participants using recombinant XBB.1.5 Spike protein. Using an optimized activation-induced marker assay, we quantified circulating Spike-specific CD4 + and CD8 + T cells based on antigen-stimulated expression of CD134, CD69, CD25, CD137, and/or CD107a. RESULTS Vaccination strongly induced SARS-CoV-2-specific T cells, including BA.4/5- and XBB.1.5-reactive T cells, which remained detectable over time and further increased following a fourth dose. However, responses to BA.4/5 (1.34- to 1.67-fold lower) XBB.1.5 (2.0- to 18-fold lower) were significantly reduced in magnitude compared with ancestral strain responses. CD4 + responses correlated with anti-receptor-binding domain antibodies and predicted subsequent antibody responses in seronegative individuals. Lung transplant recipients receiving prednisone and older adults displayed weaker responses. CONCLUSIONS Ancestral strain vaccination stimulates BA.4/5 and XBB.1.5-cross-reactive T cells in SOTRs, but at lower magnitudes. Antigen-specific T cells can predict future antibody responses. Our data support monitoring both humoral and cellular immunity in SOTRs to track COVID-19 vaccine immunogenicity against emerging variants.
Collapse
Affiliation(s)
- Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Sabine Ivison
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Qing Huang
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Gale Ladua
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Infection and Immunity Research Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Demitra M Yotis
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Dhiraj Mannar
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sriram Subramaniam
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Victor H Ferreira
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Deepali Kumar
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Sara Belga
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Infection and Immunity Research Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Bhardwaj P, Behera SP, Nanaware N, Zaman K, Deval H, Kant R, Kulkarni S, Kumar R, Dwivedi GR, Singh R. Phylogenetic and immunological investigations of complete TSA56 ORF of Orientia tsutsugamushi present in acute encephalitis syndrome cases from eastern Uttar Pradesh, India. Arch Microbiol 2023; 205:178. [PMID: 37029825 PMCID: PMC10082565 DOI: 10.1007/s00203-023-03492-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/17/2023] [Indexed: 04/09/2023]
Abstract
Scrub typhus (ST) caused by Orientia tsutsugamushi (OT), has long been known to cause acute encephalitis syndrome (AES) and acute febrile illness (AFI). The immunodominant 56 kDa protein of OT, which is encoded by the 56 kDa gene (1600 bp encoding 516-541 amino acids) is a commonly studied antigen for genotype and serotype assignment. Previous studies from India have utilized partial type specific antigen (TSA) 56 kDa sequences for OT strain characterisation. On the other hand, understanding the antigenic diversity of current OT strains, is critical for developing specific diagnostic tests and vaccines against ST. As a result, the current study analyses antigenic variants using the entire TSA56 ORF of OT from AES cases. Phylogenetic investigation using complete TSA56 ORF sequences revealed Karp and Gilliam were the circulating predominant strains of OT. Furthermore, Immuno-informatical analysis demonstrated that the majority of high-binding affinity CD4 TCEs against the most prevalent Indian human leukocyte antigen alleles were present in the S-VDIII/IV and S-VDIV spacer regions of TSA56 ORF. TSA56 conserved spacer is crucial for OT immunological response investigations. Further, the pathophysiological effects of spacer domains in ST require further investigation. Furthermore, the characterization of the TSA56 spacer region of the OT from different parts of India is critical for developing region-specific ST diagnostic assays and vaccines.
Collapse
Affiliation(s)
- Pooja Bhardwaj
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Sthita Pragnya Behera
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Nikita Nanaware
- ICMR-National AIDS Research Institute, Bhosari, Pune, 411026, India
| | - Kamran Zaman
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnatka, India
| | - Hirawati Deval
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Rajni Kant
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Smita Kulkarni
- ICMR-National AIDS Research Institute, Bhosari, Pune, 411026, India
| | - Rajesh Kumar
- RGSC, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Gaurav Raj Dwivedi
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Rajeev Singh
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India.
| |
Collapse
|
5
|
Zhang J, Li J, Hu K, Zhou Q, Chen X, He J, Yin S, Chi Y, Liao X, Xiao Y, Qin H, Zheng Z, Chen J. Screening Novel Vaccine Candidates for Leishmania Donovani by Combining Differential Proteomics and Immunoinformatics Analysis. Front Immunol 2022; 13:902066. [PMID: 35812381 PMCID: PMC9260594 DOI: 10.3389/fimmu.2022.902066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Visceral leishmaniasis (VL), also known as kala-azar, is the most dangerous form of leishmaniasis. Currently no effective vaccine is available for clinical use. Since the pathogenicity of different Leishmania strains is inconsistent, the differentially expressed proteins in Leishmania strains may play an important role as virulence factors in pathogenesis. Therefore, effective vaccine candidate targets may exist in the differentially expressed proteins. In this study, we used differential proteomics analysis to find the differentially expressed proteins in two Leishmania donovani strains, and combined with immunoinformatics analysis to find new vaccine candidates. The differentially expressed proteins from L. DD8 (low virulent) and L. 9044 (virulent) strains were analyzed by LC-MS/MS, and preliminarily screened by antigenicity, allergenicity and homology evaluation. The binding peptides of MHC II, IFN-γ and MHC I from differentially expressed proteins were then predicted and calculated for the second screening. IFN-γ/IL-10 ratios and conserved domain prediction were performed to choose more desirable differentially expressed proteins. Finally, the 3D structures of three vaccine candidate proteins were produced and submitted for molecular dynamics simulation and molecular docking interaction with TLR4/MD2. The results showed that 396 differentially expressed proteins were identified by LC-MS/MS, and 155 differentially expressed proteins were selected through antigenicity, allergenicity and homology evaluation. Finally, 16 proteins whose percentages of MHC II, IFN-γ and MHC I binding peptides were greater than those of control groups (TSA, LmSTI1, LeIF, Leish-111f) were considered to be suitable vaccine candidates. Among the 16 candidates, amino acid permease, amastin-like protein and the hypothetical protein (XP_003865405.1) simultaneously had the large ratios of IFN-γ/IL-10 and high percentages of MHC II, IFN-γ and MHC I, which should be focused on. In conclusion, our comprehensive work provided a methodological basis to screen new vaccine candidates for a better intervention against VL and associated diseases.
Collapse
Affiliation(s)
- Jianhui Zhang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiao Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Kaifeng Hu
- Department of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoxiao Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jinlei He
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Shuangshuang Yin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yangjian Chi
- Department of Urinary Surgery, Jianou Municipal Hospital of Fujian Province, Jianou, China
| | - Xuechun Liao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuying Xiao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Hanxiao Qin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhiwan Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Zhiwan Zheng, ; Jianping Chen,
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
- *Correspondence: Zhiwan Zheng, ; Jianping Chen,
| |
Collapse
|
6
|
Rostaminia S, Aghaei SS, Farahmand B, Nazari R, Ghaemi A. Computational Design and Analysis of a Multi-epitope Against Influenza A virus. Int J Pept Res Ther 2021; 27:2625-2638. [PMID: 34539293 PMCID: PMC8435298 DOI: 10.1007/s10989-021-10278-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/28/2022]
Abstract
Influenza A viruses are among the most studied viruses, however no effective prevention against influenza infection has been developed. So, designing an effective vaccine against Influenza A virus is a critical issue in the field of medical biotechnology. For this reason, to combat this disease, we have designed a novel multi-epitope vaccine candidate based on the several conserved and potential linear B-cell and T-cell binding epitopes by using the in silico approach. This vaccine consists of an ER signal conserved sequence, the PADRE conserved epitope and two conserved epitopes of Influenza matrix protein 2. T-cell binding epitopes from Matrix protein 2 were predicted by in silico tools of epitope prediction. The selected epitopes were joined by flexible linkers and physicochemical properties, toxicity, and allergenecity were investigated. The designed vaccine was antigenic, immunogenic, and non-allergenic with suitable physicochemical properties and has higher solubility. The final multi-epitope construct was modeled, confirmed by different programs and the molecular interactions with immune receptors were considered. The molecular docking assay indicated the interactions with immune-stimulatory toll-like receptor 3 (TLR3) and major histocompatibility complex class I (MHCI). The HADDOCK and H DOCK servers were used to make docking analysis, respectively. The docking analysis indicated a strong and stable binding interaction between the vaccine construct with major histocompatibility complex (MHC) class I and toll-like receptor 3. Overall, the findings suggest that the current vaccine may be a promising vaccine to prevent Influenza infection.
Collapse
Affiliation(s)
- Samaneh Rostaminia
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | | | - Behrokh Farahmand
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, 69, P.O.Box: 1316943551, Tehran, Iran
| | - Raziye Nazari
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, 69, P.O.Box: 1316943551, Tehran, Iran
| |
Collapse
|
7
|
Gorris ME, Caballero Van Dyke MC, Carey A, Hamm PS, Mead HL, Uehling JK. A Review of Coccidioides Research, Outstanding Questions in the Field, and Contributions by Women Scientists. CURRENT CLINICAL MICROBIOLOGY REPORTS 2021; 8:114-128. [PMID: 34367880 PMCID: PMC8327307 DOI: 10.1007/s40588-021-00173-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 12/19/2022]
Abstract
Purpose of Review Coccidioidomycosis is an infectious disease that gained clinical significance in the early 20th century. Many of the foundational contributions to coccidioidomycosis research, including the discovery of the fungal disease agent, Coccidioides spp., were made by women. We review recent progress in Coccidioides research and big questions remaining in the field, while highlighting some of the contributions from women. Recent Findings New molecular-based techniques provide a promising method for detecting Coccidioides, which can help determine the dominate reservoir host and ideal environmental conditions for growth. Genetic and genomic analyses have allowed an understanding of population structure, species level diversity, and evolutionary histories. We present a current, comprehensive genome list, where women contributed many of these entries. Several efforts to develop a coccidioidomycosis vaccine are underway. Summary Women continue to pioneer research on Coccidioides, including the relationships between the fungi and the environment, genetics, and clinical observations. Significant questions remain in the field of Coccidioides, including the main host reservoir, the relationships between genotypic and phenotypic variation, and the underlying cause for chronic clinical coccidioidomycosis cases.
Collapse
Affiliation(s)
- Morgan E Gorris
- Los Alamos National Laboratory, Information Systems and Modeling & Center for Nonlinear Studies, Los Alamos, NM USA
| | | | - Adrienne Carey
- Division of Infectious Diseases, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Paris S Hamm
- Department of Biology, University of New Mexico, Albuquerque, NM USA
| | - Heather L Mead
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ USA
| | - Jessie K Uehling
- Department of Botany and Plant Pathology, Oregon State University, Corvallis, OR USA
| |
Collapse
|
8
|
Noe AR, Terry FE, Schanen BC, Sassano E, Hindocha P, Phares TW, Moise L, Christen JM, Tucker KD, Kotraiah V, Drake DR, Martin WD, De Groot AS, Gutierrez GM. Bridging Computational Vaccinology and Vaccine Development Through Systematic Identification, Characterization, and Downselection of Conserved and Variable Circumsporozoite Protein CD4 T Cell Epitopes From Diverse Plasmodium falciparum Strains. Front Immunol 2021; 12:689920. [PMID: 34168657 PMCID: PMC8217813 DOI: 10.3389/fimmu.2021.689920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
An effective malaria vaccine must prevent disease in a range of populations living in regions with vastly different transmission rates and protect against genetically-diverse Plasmodium falciparum (Pf) strains. The protective efficacy afforded by the currently licensed malaria vaccine, Mosquirix™, promotes strong humoral responses to Pf circumsporozoite protein (CSP) 3D7 but protection is limited in duration and by strain variation. Helper CD4 T cells are central to development of protective immune responses, playing roles in B cell activation and maturation processes, cytokine production, and stimulation of effector T cells. Therefore, we took advantage of recent in silico modeling advances to predict and analyze human leukocyte antigen (HLA)-restricted class II epitopes from PfCSP – across the entire PfCSP 3D7 sequence as well as in 539 PfCSP sequence variants – with the goal of improving PfCSP-based malaria vaccines. Specifically, we developed a systematic workflow to identify peptide sequences capable of binding HLA-DR in a context relevant to achieving broad human population coverage utilizing cognate T cell help and with limited T regulatory cell activation triggers. Through this workflow, we identified seven predicted class II epitope clusters in the N- and C-terminal regions of PfCSP 3D7 and an additional eight clusters through comparative analysis of 539 PfCSP sequence variants. A subset of these predicted class II epitope clusters was synthesized as peptides and assessed for HLA-DR binding in vitro. Further, we characterized the functional capacity of these peptides to prime and activate human peripheral blood mononuclear cells (PBMCs), by monitoring cytokine response profiles using MIMIC® technology (Modular IMmune In vitro Construct). Utilizing this decision framework, we found sufficient differential cellular activation and cytokine profiles among HLA-DR-matched PBMC donors to downselect class II epitope clusters for inclusion in a vaccine targeting PfCSP. Importantly, the downselected clusters are not highly conserved across PfCSP variants but rather, they overlap a hypervariable region (TH2R) in the C-terminus of the protein. We recommend assessing these class II epitope clusters within the context of a PfCSP vaccine, employing a test system capable of measuring immunogenicity across a broad set of HLA-DR alleles.
Collapse
Affiliation(s)
- Amy R Noe
- Leidos Life Sciences, Leidos Inc., Frederick, MD, United States
| | | | - Brian C Schanen
- Sanofi Pasteur, VaxDesign Campus, Orlando, FL, United States
| | - Emily Sassano
- Sanofi Pasteur, VaxDesign Campus, Orlando, FL, United States
| | | | | | | | | | | | | | - Donald R Drake
- Sanofi Pasteur, VaxDesign Campus, Orlando, FL, United States
| | | | - Anne S De Groot
- EpiVax Inc., Providence, RI, United States.,University of Georgia Center for Vaccines and Immunology, Athens, GA, United States
| | | |
Collapse
|
9
|
Olotu FA, Soliman MES. Immunoinformatics prediction of potential B-cell and T-cell epitopes as effective vaccine candidates for eliciting immunogenic responses against Epstein-Barr virus. Biomed J 2021; 44:317-337. [PMID: 34154948 PMCID: PMC8358216 DOI: 10.1016/j.bj.2020.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The ongoing search for viable treatment options to curtail Epstein Barr Virus (EBV) pathogenicity has necessitated a paradigmatic shift towards the design of peptide-based vaccines. Potential B-cell and T-cell epitopes were predicted for nine antigenic EBV proteins that mediate epithelial cell-attachment and spread, capsid self-assembly, DNA replication and processivity. METHODS Predictive algorithms incorporated in the Immune Epitope Database (IEDB) resources were used to determine potential B-cell epitopes based on their physicochemical attributes. These were combined with a string-kernel method and an antigenicity predictive AlgPred tool to enhance accuracy in the end-point selection of highly potential antigenic EBV B-cell epitopes. NetCTL 1.2 algorithms enabled the prediction of probable T-cell epitopes which were structurally modeled and subjected to blind peptide-protein docking with HLA-A*02:01. All-atom molecular dynamics (MD) simulation and Molecular Mechanics Generalized-Born Surface Area methods were used to investigate interaction dynamics and affinities of predicted T-cell peptide-protein complexes. RESULTS Computational predictions and sequence overlapping analysis yielded 18 linear (continuous) and discontinuous (conformational) subunit epitopes from the antigenic proteins with characteristic surface accessibility, flexibility and antigenicity, and predictive scores above the threshold value (1) set. A novel site was identified on HLA-A*02:01 with preferential affinity binding for modeled BMRF2, BXLF1 and BGLF4 T-cell epitopes. Interaction dynamics and energies were also computed in addition to crucial residues that mediated complex formation and stability. CONCLUSION This study implemented an integrative meta-analytical approach to model highly probable B-cell and T-cell epitopes as potential peptide-vaccine candidates for the treatment of EBV-related diseases.
Collapse
Affiliation(s)
- Fisayo A Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa.
| |
Collapse
|
10
|
Forner M, Cañas-Arranz R, Defaus S, de León P, Rodríguez-Pulido M, Ganges L, Blanco E, Sobrino F, Andreu D. Peptide-Based Vaccines: Foot-and-Mouth Disease Virus, a Paradigm in Animal Health. Vaccines (Basel) 2021; 9:vaccines9050477. [PMID: 34066901 PMCID: PMC8150788 DOI: 10.3390/vaccines9050477] [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: 03/17/2021] [Revised: 04/19/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines are considered one of the greatest global health achievements, improving the welfare of society by saving lives and substantially reducing the burden of infectious diseases. However, few vaccines are fully effective, for reasons ranging from intrinsic limitations to more contingent shortcomings related, e.g., to cold chain transport, handling and storage. In this context, subunit vaccines where the essential antigenic traits (but not the entire pathogen) are presented in rationally designed fashion have emerged as an attractive alternative to conventional ones. In particular, this includes the option of fully synthetic peptide vaccines able to mimic well-defined B- and T-cell epitopes from the infectious agent and to induce protection against it. Although, in general, linear peptides have been associated to low immunogenicity and partial protection, there are several strategies to address such issues. In this review, we report the progress towards the development of peptide-based vaccines against foot-and-mouth disease (FMD) a highly transmissible, economically devastating animal disease. Starting from preliminary experiments using single linear B-cell epitopes, recent research has led to more complex and successful second-generation vaccines featuring peptide dendrimers containing multiple copies of B- and T-cell epitopes against FMD virus or classical swine fever virus (CSFV). The usefulness of this strategy to prevent other animal and human diseases is discussed.
Collapse
Affiliation(s)
- Mar Forner
- Departament de Ciències Experimentals i de la Salut (DCEXS-UPF), 08003 Barcelona, Spain; (M.F.); (S.D.)
| | - Rodrigo Cañas-Arranz
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (R.C.-A.); (P.d.L.); (M.R.-P.)
| | - Sira Defaus
- Departament de Ciències Experimentals i de la Salut (DCEXS-UPF), 08003 Barcelona, Spain; (M.F.); (S.D.)
| | - Patricia de León
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (R.C.-A.); (P.d.L.); (M.R.-P.)
| | - Miguel Rodríguez-Pulido
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (R.C.-A.); (P.d.L.); (M.R.-P.)
| | - Llilianne Ganges
- Centre de Recerca en Sanitat Animal (CReSA), OIE Reference Laboratory for Classical Swine Fever, Institute of Agrifood Research and Technology, 08193 Barcelona, Spain;
| | - Esther Blanco
- Centro de Investigación en Sanidad Animal (CISA-INIA), 28130 Valdeolmos, Spain;
| | - Francisco Sobrino
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (R.C.-A.); (P.d.L.); (M.R.-P.)
- Correspondence: (F.S.); (D.A.)
| | - David Andreu
- Departament de Ciències Experimentals i de la Salut (DCEXS-UPF), 08003 Barcelona, Spain; (M.F.); (S.D.)
- Correspondence: (F.S.); (D.A.)
| |
Collapse
|
11
|
Dalvie NC, Brady JR, Crowell LE, Tracey MK, Biedermann AM, Kaur K, Hickey JM, Kristensen DL, Bonnyman AD, Rodriguez-Aponte SA, Whittaker CA, Bok M, Vega C, Mukhopadhyay TK, Joshi SB, Volkin DB, Parreño V, Love KR, Love JC. Molecular engineering improves antigen quality and enables integrated manufacturing of a trivalent subunit vaccine candidate for rotavirus. Microb Cell Fact 2021; 20:94. [PMID: 33933073 PMCID: PMC8088319 DOI: 10.1186/s12934-021-01583-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Vaccines comprising recombinant subunit proteins are well-suited to low-cost and high-volume production for global use. The design of manufacturing processes to produce subunit vaccines depends, however, on the inherent biophysical traits presented by an individual antigen of interest. New candidate antigens typically require developing custom processes for each one and may require unique steps to ensure sufficient yields without product-related variants. RESULTS We describe a holistic approach for the molecular design of recombinant protein antigens-considering both their manufacturability and antigenicity-informed by bioinformatic analyses such as RNA-seq, ribosome profiling, and sequence-based prediction tools. We demonstrate this approach by engineering the product sequences of a trivalent non-replicating rotavirus vaccine (NRRV) candidate to improve titers and mitigate product variants caused by N-terminal truncation, hypermannosylation, and aggregation. The three engineered NRRV antigens retained their original antigenicity and immunogenicity, while their improved manufacturability enabled concomitant production and purification of all three serotypes in a single, end-to-end perfusion-based process using the biotechnical yeast Komagataella phaffii. CONCLUSIONS This study demonstrates that molecular engineering of subunit antigens using advanced genomic methods can facilitate their manufacturing in continuous production. Such capabilities have potential to lower the cost and volumetric requirements in manufacturing vaccines based on recombinant protein subunits.
Collapse
Affiliation(s)
- Neil C Dalvie
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Joseph R Brady
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Laura E Crowell
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mary Kate Tracey
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Andrew M Biedermann
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kawaljit Kaur
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, 66047, USA
| | - John M Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, 66047, USA
| | - D Lee Kristensen
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Alexandra D Bonnyman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sergio A Rodriguez-Aponte
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Charles A Whittaker
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Marina Bok
- Instituto de Virología E Innovaciones Tecnológicas, IVIT, CONICET-INTA, Hurlingham,, Buenos Aires, Argentina
| | - Celina Vega
- Instituto de Virología E Innovaciones Tecnológicas, IVIT, CONICET-INTA, Hurlingham,, Buenos Aires, Argentina
| | - Tarit K Mukhopadhyay
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, 66047, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, 66047, USA
| | - Viviana Parreño
- Instituto de Virología E Innovaciones Tecnológicas, IVIT, CONICET-INTA, Hurlingham,, Buenos Aires, Argentina
| | - Kerry R Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - J Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
12
|
Mahmoudvand S, Shokri S, Makvandi M, Taherkhani R, Rashno M, Jalilian FA, Angali KA. In silico prediction of T-cell and B-cell epitopes of human papillomavirus type 16 L1 protein. Biotechnol Appl Biochem 2021; 69:514-525. [PMID: 33624357 DOI: 10.1002/bab.2128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Human papillomavirus type 16 (HPV-16) is one of the most important cause of developing cervical cancer. Therefore, effective epitope-based vaccine design for HPV-16 would be of major medical benefit. The aim of our study was to identify B- and T-cell epitopes of HPV-16 L1 protein. In this study, the HPV-16 L1 gene was isolated from HPV recovered from five vaginal swab samples using specific primers and finally sequenced. The ExPASy translate tool (http://web.expasy.org/translate/) was used to convert nucleotide sequence into amino acid sequence. Bioinformatic analysis was employed to predict suitable B- and T-cell epitopes and immunogenicity, allergenicity, and toxicity of predicted epitopes were then evaluated. Afterward, the selected T-cell epitopes were docked using Molegro Virtual Docker software. The two epitopes 207 AMDFTTLQA215 and 200 MVDTGFGAM208 have showed a very strong binding affinity to HLA-A0201 and HLA-B3501 molecules, respectively. Outcome of B-cell epitope prediction showed that epitope 475 KAKPKFTLGKRK ATPTTSSTSTTAKRKK502 contained overlapped epitope, which might be the epitope associated with the production of neutralizing antibody response. Based on this finding, the predicted B- and T-cell epitopes are promising targets for epitope-based vaccine development against HPV-16. Further in vivo and in vitro experiments are needed to confirm our findings.
Collapse
Affiliation(s)
- Shahab Mahmoudvand
- Infectious and Tropical Disease Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Shokri
- Infectious and Tropical Disease Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manoochehr Makvandi
- Infectious and Tropical Disease Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Taherkhani
- Persian Gulf Biomedical Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Rashno
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farid Azizi Jalilian
- Department of Virology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Kambiz Ahmadi Angali
- Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
13
|
Namazi F, Davoodi S, Bolhassani A. Comparison of the efficacy of HIV-1 Nef-Tat-Gp160-p24 polyepitope vaccine candidate with Nef protein in different immunization strategies. Curr Drug Deliv 2021; 19:142-156. [PMID: 33655833 DOI: 10.2174/1567201818666210224101144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/23/2020] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVES One of the promising strategies for effective HIV-1 vaccine design involves finding the polyepitope immunogens using T cell epitopes. METHODS Herein, an HIV-1 polyepitope construct (i.e., Nef-Tat-Gp160-P24) comprising of several epitopes from Nef, Tat, Gp160, and P24 proteins was designed. To improve its immunogenicity in BALB/c mice, cell-penetrating peptides (HR9 & MPG for DNA delivery, and LDP-NLS & CyLoP-1 for protein transfer), Montanide adjuvant, and heterologous DNA prime/polypeptide boost strategy were used. To compare the immunogenicity, Nef was utilized as a vaccine candidate. The levels of total IgG and its subclasses, cytokines, and Granzyme B were assessed using ELISA. RESULTS Immunological studies showed that heterologous prime-boost regimens for both antigens could considerably augment the levels of IgG2a, IgG2b, IFN-γ, and Granzyme B directed toward Th1 and CTL immune responses in comparison with homologous prime-boost strategies. The levels of IFN-γ, IL-10, total IgG, IgG1, and IgG2b were drastically higher in groups immunized with Nef-Tat-Gp160-P24 in heterologous prime-boost regimens than those in groups immunized with Nef. CONCLUSIONS The use of the Nef-Tat-Gp160-P24 polyepitope immunogen in heterologous prime-boost strategy could generate the mixture of Th1 and Th2 responses directed further toward Th1 response as a hopeful method for improvement of HIV-1 vaccine.
Collapse
Affiliation(s)
- Fatemeh Namazi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran. Iran
| | - Saba Davoodi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran. Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran. Iran
| |
Collapse
|
14
|
Mahdevar E, Safavi A, Abiri A, Kefayat A, Hejazi SH, Miresmaeili SM, Iranpur Mobarakeh V. Exploring the cancer-testis antigen BORIS to design a novel multi-epitope vaccine against breast cancer based on immunoinformatics approaches. J Biomol Struct Dyn 2021; 40:6363-6380. [PMID: 33599191 DOI: 10.1080/07391102.2021.1883111] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recently, cancer immunotherapy has gained lots of attention to replace the current chemoradiation approaches and multi-epitope cancer vaccines are manifesting as the next generation of cancer immunotherapy. Therefore, in this study, we used multiple immunoinformatics approaches along with other computational approaches to design a novel multi-epitope vaccine against breast cancer. The most immunogenic regions of the BORIS cancer-testis antigen were selected according to the binding affinity to MHC-I and II molecules as well as containing multiple cytotoxic T lymphocyte (CTL) epitopes by multiple immunoinformatics servers. The selected regions were linked together by GPGPG linker. Also, a T helper epitope (PADRE) and the TLR-4/MD-2 agonist (L7/L12 ribosomal protein from mycobacterium) were incorporated by A(EAAAK)3A linker to form the final vaccine construct. Then, its physicochemical properties, cleavage sites, TAP transport efficiency, B cell epitopes, IFN-γ inducing epitopes and population coverage were predicted. The final vaccine construct was reverse translated, codon-optimized and inserted into pcDNA3.1 to form the DNA vaccine. The final vaccine construct was a stable, immunogenic and non-allergenic protein that contained numerous CTL epitopes, IFN-γ inducing epitopes and several linear and conformational B cell epitopes. Also, the final vaccine construct formed stable and significant interactions with TLR-4/MD-2 complex according to molecular docking and dynamics simulations. Moreover, its world population coverage for HLA-I and HLA-II were about 93% and 96%, respectively. Taking together, these preliminary results can be used as an appropriate platform for further experimental investigations.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Elham Mahdevar
- Department of Biology, Faculty of Science and Engineering, Science and Arts University, Yazd, Iran
| | - Ashkan Safavi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ardavan Abiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Hossein Hejazi
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Mohsen Miresmaeili
- Department of Biology, Faculty of Science and Engineering, Science and Arts University, Yazd, Iran
| | | |
Collapse
|
15
|
Liu J, Chen X, Wang J, Wu F, Zhang J, Dong J, Zhang H, Liu X, Hu N, Wu J, Zhang L, Cheng W, Zhang C, Zhang WJ. Prediction and identification of CD4+ T cell epitope for the protective antigens of Mycobacterium tuberculosis. Medicine (Baltimore) 2021; 100:e24619. [PMID: 33578573 PMCID: PMC7886468 DOI: 10.1097/md.0000000000024619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/08/2021] [Indexed: 02/05/2023] Open
Abstract
CD4+T cell epitopes plays a key role in anti-tuberculosis (TB) immunity, CD4+T cell epitopes suitable for the domestic population are lacking. Therefore, we predicted and identified novel CD4+T cell epitopes.The bioinformatics software, namely, DNAStar (DNASTAR of the United States), SYFPEITHI (INTERFACTORS INSTITUT Für ZELL Biologie of Germany), RANKPEP, and NetMHC IIpan (National Cancer Institute, United States of America), were used to comprehensively predict the CD4+T cell immune epitope of Mycobacterium TB, and the predicted epitope polypeptide was synthesized by the standard Fmoc scheme. The proliferation of PBMC and CD4+T cells stimulated by peptides was preliminarily detected by the CCK8 method. Then, the candidate polypeptides screened out by the CCK8 method were verified again by the BrdU assay, and flow cytometry was performed to analyze further the extent of their stimulation on the proliferation of CD4+T cells. The changes in the secreted cytokines IFN-γ, TNF-α, IL-2, and IL-10 before and after the candidate polypeptide stimulation of CD4+T lymphocytes were detected by ELISA. The preliminary humoral immunity test was conducted by indirect ELISA to evaluate the serological diagnostic value of the CD4+T cell epitope polypeptide.In this study, 5 novel candidate CD4+T cell epitope polypeptides with the amino acid sequences of LQGQWRGAAGTAAQA, PVTLAETGSTLLYPL, AAAWGGSGSEAYQGV, QFVYAGAMSGLLDPS, and KAALTRTASNMNAAA and others that have not been reported in the research were predicted. For convenience, the 5 candidates were successively named as P39, P50, P40, P185, and P62. P39, P62, and the mixed peptide P39+P62 could effectively induce the proliferation of CD4+T cells and increase the secretion of IFN-γ, TNF-α, and IL-2 from the CD4+T cells, while reducing the content of IL-10. The serological test showed that the sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) of P39 were 75%, 67.71%, and 0.844, respectively. The sensitivity, specificity, and AUC of P62 were 91.66%, 46.87%, and 0.649, respectively. The sensitivity of the mixed peptide P39+P62 was 95.83%, the specificity was 97.91%, and the AUC was 0.793.The P39 and P62 polypeptides were predicted and identified as potential CD4+T cell immune epitope polypeptides of M. TB. The polypeptide had better diagnosis effect, which provided potential candidate epitope polypeptides for the development of TB-specific diagnosis reagents and novel TB epitope vaccines.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Xuefeng Chen
- West China Hospital of Sichuan University, Wuhou District, Chengdu, Sichuan
| | - Ju Wang
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Fang Wu
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Jie Zhang
- The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, P. R. China
| | - Jiangtao Dong
- The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, P. R. China
| | - Hui Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Xiaoling Liu
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Na Hu
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Jiangdong Wu
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Le Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Wei Cheng
- West China Hospital of Sichuan University, Wuhou District, Chengdu, Sichuan
| | - Chunjun Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| | - Wan Jiang Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/the Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang
| |
Collapse
|
16
|
A comprehensive screening of the whole proteome of hantavirus and designing a multi-epitope subunit vaccine for cross-protection against hantavirus: Structural vaccinology and immunoinformatics study. Microb Pathog 2020; 150:104705. [PMID: 33352214 DOI: 10.1016/j.micpath.2020.104705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/18/2020] [Accepted: 12/10/2020] [Indexed: 11/23/2022]
Abstract
Hantaviruses are an emerging zoonotic group of rodent-borne viruses that are having serious implications on global public health due to the increase in outbreaks. Since there is no permanent cure, there is increasing interest in developing a vaccine against the hantavirus. This research aimed to design a robust cross-protective subunit vaccine using a novel immunoinformatics approach. After careful evaluation, the best predicted cytotoxic & helper T-cell and B-cell epitopes from nucleocapsid proteins, glycoproteins, RdRp proteins, and non-structural proteins were considered as potential vaccine candidates. Among the four generated vaccine models with different adjuvant, the model with toll-like receptor-4 (TLR-4) agonist adjuvant was selected because of its high antigenicity, non-allergenicity, and structural quality. The selected model was 654 amino acids long and had a molecular weight of 70.5 kDa, which characterizes the construct as a good antigenic vaccine candidate. The prediction of the conformational B-lymphocyte (CBL) epitope secured its ability to induce the humoral response. Thereafter, disulfide engineering improved vaccine stability. Afterwards, the molecular docking confirmed a good binding affinity of -1292 kj/mol with considered immune receptor TLR-4 and the dynamics simulation showed high stability of the vaccine-receptor complex. Later, the in silico cloning confirmed the better expression of the constructed vaccine protein in E. coli K12. Finally, in in silico immune simulation, significantly high levels of immunoglobulin M (IgM), immunoglobulin G1 (IgG1), cytotoxic & helper T lymphocyte (CTL & HTL) populations, and numerous cytokines such as interferon-γ (IFN-γ), interleukin-2 (IL-2) etc. were found as coherence with actual immune response and also showed faster antigen clearance for repeated exposures. Nonetheless, experimental validation can demonstrate the safety and cross-protective ability of the proposed vaccine to fight against hantavirus infection.
Collapse
|
17
|
Tilocca B, Britti D, Urbani A, Roncada P. Computational Immune Proteomics Approach to Target COVID-19. J Proteome Res 2020; 19:4233-4241. [PMID: 32914632 PMCID: PMC7640973 DOI: 10.1021/acs.jproteome.0c00553] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 12/28/2022]
Abstract
Progress of the omics platforms widens their application to diverse fields, including immunology. This enables a deeper level of knowledge and the provision of a huge amount of data for which management and fruitful integration with the past evidence requires a steadily growing computational effort. In light of this, immunoinformatics emerges as a new discipline placed in between the traditional lab-based investigations and the computational analysis of the biological data. Immunoinformatics make use of tailored bioinformatics tools and data repositories to facilitate the analysis of data from a plurality of disciplines and help drive novel research hypotheses and in silico screening investigations in a fast, reliable, and cost-effective manner. Such computational immunoproteomics studies may as well prepare and guide lab-based investigations, representing valuable technology for the investigation of novel pathogens, to tentatively evaluate specificity of diagnostic products, to forecast on potential adverse effects of vaccines and to reduce the use of animal models. The present manuscript provides an overview of the COVID-19 pandemic and reviews the state of the art of the omics technologies employed in fighting SARS-CoV-2 infections. A comprehensive description of the immunoinformatics approaches and its potential role in contrasting COVID-19 pandemics is provided.
Collapse
Affiliation(s)
- Bruno Tilocca
- Department
of Health Sciences, University “Magna
Graecia” of Catanzaro, Catanzaro 88100, Italy
| | - Domenico Britti
- Department
of Health Sciences, University “Magna
Graecia” of Catanzaro, Catanzaro 88100, Italy
| | - Andrea Urbani
- Department
of Basic Biotechnological Sciences, Intensivological and Perioperative
Clinics, Università Cattolica del
Sacro Cuore, Roma 00168, Italy
- Dipartimento
di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario Agostino Gemelli, Roma 00168, Italy
| | - Paola Roncada
- Department
of Health Sciences, University “Magna
Graecia” of Catanzaro, Catanzaro 88100, Italy
| |
Collapse
|
18
|
Rakib A, Sami SA, Mimi NJ, Chowdhury MM, Eva TA, Nainu F, Paul A, Shahriar A, Tareq AM, Emon NU, Chakraborty S, Shil S, Mily SJ, Ben Hadda T, Almalki FA, Emran TB. Immunoinformatics-guided design of an epitope-based vaccine against severe acute respiratory syndrome coronavirus 2 spike glycoprotein. Comput Biol Med 2020; 124:103967. [PMID: 32828069 PMCID: PMC7423576 DOI: 10.1016/j.compbiomed.2020.103967] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/08/2020] [Accepted: 08/08/2020] [Indexed: 02/07/2023]
Abstract
AIMS With a large number of fatalities, coronavirus disease-2019 (COVID-19) has greatly affected human health worldwide. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus that causes COVID-19. The World Health Organization has declared a global pandemic of this contagious disease. Researchers across the world are collaborating in a quest for remedies to combat this deadly virus. It has recently been demonstrated that the spike glycoprotein (SGP) of SARS-CoV-2 is the mediator by which the virus enters host cells. MAIN METHODS Our group comprehensibly analyzed the SGP of SARS-CoV-2 through multiple sequence analysis and a phylogenetic analysis. We predicted the strongest immunogenic epitopes of the SGP for both B cells and T cells. KEY FINDINGS We focused on predicting peptides that would bind major histocompatibility complex class I. Two optimal epitopes were identified, WTAGAAAYY and GAAAYYVGY. They interact with the HLA-B*15:01 allele, which was further validated by molecular docking simulation. This study also found that the selected epitopes are able to be recognized in a large percentage of the world's population. Furthermore, we predicted CD4+ T-cell epitopes and B-cell epitopes. SIGNIFICANCE Our study provides a strong basis for designing vaccine candidates against SARS-CoV-2. However, laboratory work is required to validate our theoretical results, which would lay the foundation for the appropriate vaccine manufacturing and testing processes.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Betacoronavirus/genetics
- Betacoronavirus/immunology
- COVID-19
- COVID-19 Vaccines
- Computational Biology
- Coronavirus Infections/epidemiology
- Coronavirus Infections/genetics
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Drug Design
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- HLA-B15 Antigen/chemistry
- HLA-B15 Antigen/metabolism
- HLA-DRB1 Chains/chemistry
- HLA-DRB1 Chains/metabolism
- Humans
- Molecular Docking Simulation
- Pandemics/prevention & control
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/immunology
- Viral Vaccines/chemistry
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Ahmed Rakib
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Saad Ahmed Sami
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Nusrat Jahan Mimi
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Md Mustafiz Chowdhury
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Taslima Akter Eva
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Kota Makassar, Sulawesi Selatan, 90245, Indonesia
| | - Arkajyoti Paul
- Drug Discovery, GUSTO A Research Group, Chittagong, 4203, Bangladesh; Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Asif Shahriar
- Department of Microbiology, Stamford University Bangladesh, 51 Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Abu Montakim Tareq
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh
| | - Nazim Uddin Emon
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh
| | - Sajal Chakraborty
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Sagar Shil
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Sabrina Jahan Mily
- Department of Gynaecology and Obstetrics, Banshkhali Upazila Health Complex, Jaldi Union, Chittagong, 4390, Bangladesh
| | - Taibi Ben Hadda
- Laboratory of Applied Chemistry & Environment, Faculty of Sciences, University Mohammed the First, BP 524, 60000, Oujda, Morocco; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah Almukkarramah, 21955, Saudi Arabia.
| | - Faisal A Almalki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah Almukkarramah, 21955, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.
| |
Collapse
|
19
|
Bartlett S, Skwarczynski M, Toth I. Lipids as Activators of Innate Immunity in Peptide Vaccine Delivery. Curr Med Chem 2020; 27:2887-2901. [PMID: 30362416 DOI: 10.2174/0929867325666181026100849] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 05/16/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Innate immune system plays an important role in pathogen detection and the recognition of vaccines, mainly through pattern recognition receptors (PRRs) that identify pathogen components (danger signals). One of the typically recognised bacterial components are lipids in conjugation with peptides, proteins and saccharides. Lipidic compounds are readily recognised by the immune system, and thus are ideal candidates for peptide- based vaccine delivery. Thus, bacterial or synthetic lipids mixed with, or conjugated to, antigens have shown adjuvant properties. These systems have many advantages over traditional adjuvants, including low toxicity and good efficacy for stimulating mucosal and systemic immune responses. METHODS The most recent literature on the role of lipids in stimulation of immune responses was selected for this review. The vast majority of reviewed papers were published in the last decade. Older but significant findings are also cited. RESULTS This review focuses on the development of lipopeptide vaccine systems including application of palmitic acid, bacterial lipopeptides, glycolipids and the lipid core peptide and their routes of administration. The use of liposomes as a delivery system that incorporates lipopeptides is discussed. The review also includes a brief description of immune system in relation to vaccinology and discussion on vaccine delivery routes. CONCLUSION Lipids and their conjugates are an ideal frontrunner in the development of safe and efficient vaccines for different immunisation routes.
Collapse
Affiliation(s)
- Stacey Bartlett
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD, 4072, Australia
| | - Mariusz Skwarczynski
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD, 4072, Australia
| | - Istvan Toth
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD, 4072, Australia.,The University of Queensland, School of Pharmacy, Woolloongabba, QLD 4102, Australia.,The University of Queensland, Institute for Molecular Bioscience, St Lucia, QLD 4072, Australia
| |
Collapse
|
20
|
Choga WT, Anderson M, Zumbika E, Phinius BB, Mbangiwa T, Bhebhe LN, Baruti K, Kimathi PO, Seatla KK, Musonda RM, Bell TG, Moyo S, Blackard JT, Gaseitsiwe S. In Silico Prediction of Human Leukocytes Antigen (HLA) Class II Binding Hepatitis B Virus (HBV) Peptides in Botswana. Viruses 2020; 12:E731. [PMID: 32640609 PMCID: PMC7412261 DOI: 10.3390/v12070731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B virus (HBV) is the primary cause of liver-related malignancies worldwide, and there is no effective cure for chronic HBV infection (CHB) currently. Strong immunological responses induced by T cells are associated with HBV clearance during acute infection; however, the repertoire of epitopes (epi) presented by major histocompatibility complexes (MHCs) to elicit these responses in various African populations is not well understood. In silico approaches were used to map and investigate 15-mers HBV peptides restricted to 9 HLA class II alleles with high population coverage in Botswana. Sequences from 44 HBV genotype A and 48 genotype D surface genes (PreS/S) from Botswana were used. Of the 1819 epi bindings predicted, 20.2% were strong binders (SB), and none of the putative epi bind to all the 9 alleles suggesting that multi-epitope, genotype-based, population-based vaccines will be more effective against HBV infections as opposed to previously proposed broad potency epitope-vaccines which were assumed to work for all alleles. In total, there were 297 unique epi predicted from the 3 proteins and amongst, S regions had the highest number of epi (n = 186). Epitope-densities (Depi) between genotypes A and D were similar. A number of mutations that hindered HLA-peptide binding were observed. We also identified antigenic and genotype-specific peptides with characteristics that are well suited for the development of sensitive diagnostic kits. This study identified candidate peptides that can be used for developing multi-epitope vaccines and highly sensitive diagnostic kits against HBV infection in an African population. Our results suggest that viral variability may hinder HBV peptide-MHC binding, required to initiate a cascade of immunological responses against infection.
Collapse
Affiliation(s)
- Wonderful Tatenda Choga
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Motswedi Anderson
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
| | - Edward Zumbika
- Department of Applied Biology and Biochemistry, Faculty of Applied Sciences, National University of Science and Technology, Bulawayo 0000, Zimbabwe;
| | - Bonolo B. Phinius
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
| | - Tshepiso Mbangiwa
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Lynnette N. Bhebhe
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
| | - Kabo Baruti
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Department of Biological Sciences, Faculty of Science, University of Botswana, Gaborone 0000, Botswana
| | | | - Kaelo K. Seatla
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Botswana, Gaborone 0000, Botswana
| | - Rosemary M. Musonda
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Trevor Graham Bell
- Independent Researcher, P.O. Box 497, Wits, Johannesburg 2050, South Africa;
| | - Sikhulile Moyo
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jason T. Blackard
- Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Simani Gaseitsiwe
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone 0000, Botswana; (W.T.C.); (M.A.); (B.B.P.); (T.M.); (L.N.B.); (K.B.); (K.K.S.); (R.M.M.); (S.M.)
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
21
|
Yang S, Li W, Fan Z, Zhai L, Chen J, Xiao X, Ma J, Song B, Ma J, Tong C, Yu L, Yu Y, Cao W, Cui Y. Identification of CD4 + T cell epitopes on glyceraldehyde-3-phosphate dehydrogenase-C of Staphylococcus aureus in Babl/c mice. Microb Pathog 2020; 144:104167. [PMID: 32222538 DOI: 10.1016/j.micpath.2020.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 11/15/2022]
Abstract
Glyceraldehyde-3-phosphate dehydrogenase-C (GapC) is a highly conserved surface protein of Staphylococcus aureus, with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity, which represents an excellent vaccine candidate antigen. It can induce protective immune responses to S. aureus infections. However, CD4+ T cell epitopes of GapC that induce CD4+ T cell immune responses are currently unclear. In this study, we used bioinformatics prediction algorithms to predict CD4+ T cell epitopes of GapC. Ten peptides were synthesized to investigate the candidate epitopes. Our results showed that the peptides, G4 (GapC 104-123) and G10 (GapC 314-333) were able to induce proliferation of CD4+ T cells and secrete high levels of interferon (IFN)-γ, respectively. In addition, they significantly reduced bacterial loads in tissue and induced immunoprotective effects. It is suggested that G4 and G10 are Th1-type epitopes of S. aureus GapC. This study provides the potential development of the design of epitope-based vaccine against S. aureus.
Collapse
Affiliation(s)
- Siyu Yang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Wanyu Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Zhaowei Fan
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Lu Zhai
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jing Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Xue Xiao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jun Ma
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Baifen Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Liquan Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yongzhong Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Weifan Cao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yudong Cui
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| |
Collapse
|
22
|
Abdulla F, Adhikari UK, Uddin MK. Exploring T & B-cell epitopes and designing multi-epitope subunit vaccine targeting integration step of HIV-1 lifecycle using immunoinformatics approach. Microb Pathog 2019; 137:103791. [PMID: 31606417 DOI: 10.1016/j.micpath.2019.103791] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Abstract
Till now, AIDS, caused by the human immunodeficiency virus (HIV) is still a severe health problem worldwide. It weakens the immune system by targeting the T-helper cells. Specifically, the severity of the pandemic HIV-1 makes the emergence of an enduring effective vaccine against HIV-1. Therefore, we have applied a series of immunoinformatics approaches to the four conserved domains of HIV-1 integrase (IN) proteins to design an effective multi-epitope based subunit vaccine which might induce a competent immunity against HIV-1. Therefore, we have selected three peptide fragments that contained all overlapping epitopes (35 CD4+, 8 CD8+ T-cell epitopes, and 3 B-cell epitopes) where the epitopes had a high conservancy score. The cumulative population coverage for combined CD8+ and CD4+ T-cell epitopes and their respective HLA-alleles were found as 98.03% in the world which is also followed by East Asia (96.24%), South Asia (96.31%), North Africa (96.14%), North America (98.99%), and Europe (98.80%). The proposed vaccine composed by an adjuvant (β-defensin) at the N-terminal site of the vaccine constructs and three peptide fragments where the adjuvant was fused by EAAAK linker and the peptide fragments were fused by GPGPG linker. The designed final vaccine construct (length: 159 amino acid) was found to be antigenic and non-allergic, which indicates its safety. The vaccine construct was found as good antigenic, stable, higher thermostable, and hydrophilic in nature. The codon adaptation and in silico cloning ensured the high expression rate of the vaccine constructs in E. coli K12 with CAI value of 1.0. Finally, the binding affinity of the vaccine constructs with the immune receptor TLR3 was confirmed by the lowest energy score of -1026.8 evaluated by molecular docking. However, the proposed in silico vaccine construct needs experimental validation for assuring the safety and immunogenicity profile which will ensure an active immunity against HIV-1.
Collapse
Affiliation(s)
- Faruq Abdulla
- Department of Statistics, Islamic University, Kushtia-7003, Bangladesh
| | - Utpal Kumar Adhikari
- School of Medicine, Western Sydney University, Campbelltown, NSW-2560, Australia
| | - M Kamal Uddin
- Department of Statistics, Islamic University, Kushtia-7003, Bangladesh.
| |
Collapse
|
23
|
Van Dyke MCC, Thompson GR, Galgiani JN, Barker BM. The Rise of Coccidioides: Forces Against the Dust Devil Unleashed. Front Immunol 2019; 10:2188. [PMID: 31572393 PMCID: PMC6749157 DOI: 10.3389/fimmu.2019.02188] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
Coccidioidomycosis (Valley fever) is a fungal disease caused by the inhalation of Coccidioides posadasii or C. immitis. This neglected disease occurs in the desert areas of the western United States, most notably in California and Arizona, where infections continue to rise. Clinically, coccidioidomycosis ranges from asymptomatic to severe pulmonary disease and can disseminate to the brain, skin, bones, and elsewhere. New estimates suggest as many as 350,000 new cases of coccidioidomycosis occur in the United States each year. Thus, there is an urgent need for the development of a vaccine and new therapeutic drugs against Coccidioides infection. In this review, we discuss the battle against Coccidioides including the development of potential vaccines, the quest for new therapeutic drugs, and our current understanding of the protective host immune response to Coccidioides infection.
Collapse
Affiliation(s)
| | - George R Thompson
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States.,Division of Infectious Diseases, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, United States
| | - John N Galgiani
- Valley Fever Center for Excellence, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, United States
| | - Bridget M Barker
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| |
Collapse
|
24
|
Garde C, Ramarathinam SH, Jappe EC, Nielsen M, Kringelum JV, Trolle T, Purcell AW. Improved peptide-MHC class II interaction prediction through integration of eluted ligand and peptide affinity data. Immunogenetics 2019; 71:445-454. [PMID: 31183519 DOI: 10.1007/s00251-019-01122-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/31/2019] [Indexed: 01/17/2023]
Abstract
Major histocompatibility complex (MHC) class II antigen presentation is a key component in eliciting a CD4+ T cell response. Precise prediction of peptide-MHC (pMHC) interactions has thus become a cornerstone in defining epitope candidates for rational vaccine design. Current pMHC prediction tools have, so far, primarily focused on inference from in vitro binding affinity. In the current study, we collate a large set of MHC class II eluted ligands generated by mass spectrometry to guide the prediction of MHC class II antigen presentation. We demonstrate that models developed on eluted ligands outperform those developed on pMHC binding affinity data. The predictive performance can be further enhanced by combining the eluted ligand and pMHC affinity data in a single prediction model. Furthermore, by including ligand data, the peptide length preference of MHC class II can be accurately learned by the prediction model. Finally, we demonstrate that our model significantly outperforms the current state-of-the-art prediction method, NetMHCIIpan, on an external dataset of eluted ligands and appears superior in identifying CD4+ T cell epitopes.
Collapse
Affiliation(s)
- Christian Garde
- Evaxion Biotech, Bredgade 34E, DK-1260, Copenhagen, Denmark.
| | - Sri H Ramarathinam
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Emma C Jappe
- Evaxion Biotech, Bredgade 34E, DK-1260, Copenhagen, Denmark.,Department of Bio and Health Informatics, Technical University of Denmark, DK-2800, Lyngby, Denmark
| | - Morten Nielsen
- Department of Bio and Health Informatics, Technical University of Denmark, DK-2800, Lyngby, Denmark.,Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | | | - Thomas Trolle
- Evaxion Biotech, Bredgade 34E, DK-1260, Copenhagen, Denmark
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
25
|
Panahi HA, Bolhassani A, Javadi G, Noormohammadi Z. A comprehensive in silico analysis for identification of therapeutic epitopes in HPV16, 18, 31 and 45 oncoproteins. PLoS One 2018; 13:e0205933. [PMID: 30356257 PMCID: PMC6200245 DOI: 10.1371/journal.pone.0205933] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/11/2018] [Indexed: 11/25/2022] Open
Abstract
Human papillomaviruses (HPVs) are a group of circular double-stranded DNA viruses, showing severe tropism to mucosal tissues. A subset of HPVs, especially HPV16 and 18, are the primary etiological cause for several epithelial cell malignancies, causing about 5.2% of all cancers worldwide. Due to the high prevalence and mortality, HPV-associated cancers have remained as a significant health problem in human society, making an urgent need to develop an effective therapeutic vaccine against them. Achieving this goal is primarily dependent on the identification of efficient tumor-associated epitopes, inducing a robust cell-mediated immune response. Previous information has shown that E5, E6, and E7 early proteins are responsible for the induction and maintenance of HPV-associated cancers. Therefore, the prediction of major histocompatibility complex (MHC) class I T cell epitopes of HPV16, 18, 31 and 45 oncoproteins was targeted in this study. For this purpose, a two-step plan was designed to identify the most probable CD8+ T cell epitopes. In the first step, MHC-I and II binding, MHC-I processing, MHC-I population coverage and MHC-I immunogenicity prediction analyses, and in the second step, MHC-I and II protein-peptide docking, epitope conservation, and cross-reactivity with host antigens' analyses were carried out successively by different tools. Finally, we introduced five probable CD8+ T cell epitopes for each oncoprotein of the HPV genotypes (60 epitopes in total), which obtained better scores by an integrated approach. These predicted epitopes are valuable candidates for in vitro or in vivo therapeutic vaccine studies against the HPV-associated cancers. Additionally, this two-step plan that each step includes several analyses to find appropriate epitopes provides a rational basis for DNA- or peptide-based vaccine development.
Collapse
Affiliation(s)
- Heidar Ali Panahi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Gholamreza Javadi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
26
|
Spray dried cubosomes with ovalbumin and Quil-A as a nanoparticulate dry powder vaccine formulation. Int J Pharm 2018; 550:35-44. [DOI: 10.1016/j.ijpharm.2018.08.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 01/30/2023]
|
27
|
Kalyanaraman N. In silico prediction of potential vaccine candidates on capsid protein of human bocavirus 1. Mol Immunol 2017; 93:193-205. [PMID: 29207326 DOI: 10.1016/j.molimm.2017.11.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/18/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Human bocavirus 1 (HBoV1) is a newly identified parvovirus that causes serious respiratory infection among children across the globe. Aim of the present study was to predict immunogenic residues located on the VP2 protein of HBoV1 towards development of epitope based vaccines. Several computational tools were employed to predict epitopes (bothT and B cell restricted) with stringent regulation for the improvement of confidence. After meticulous analysis, the peptide "TTPWTYFNFNQY" was identified as potential candidate for development of preventive vaccine. Of note, the epitope "TTPWTYFNFNQY" was found to be recognized by fifteen different alleles belonging to seven HLA supertypes (A1, A3, A24, A26, B7, B58 and B62). Further, mutational variability analysis pointed that most of the amino acids were well conserved. Docking scores obtained from ClusPro and Autodock Vina for selected epitopes displayed energetically favorable and stable interaction of peptide-HLA-I complexes. The core peptide "LLYQMPFFL" was found to recognize by wide range of HLA class II allele recognition thereby qualified as candidate for therapeutic vaccine. Five distinct linear peptides (withT cell epitope superimposition) belonging to B cells were identified in the VP2 protein. Further attention on the enlisted epitopes may shed light on the path for development of diagnostic, therapeutic and preventive tools against HBoV1 infection. Additionally, the predicted epitopes may help us to address the original antigenic sin phenomena observed during consecutive HBoV2-4 infection.
Collapse
Affiliation(s)
- Narayanan Kalyanaraman
- Viral Research Diagnostic Laboratory (VRDL), Department of Microbiology, Govt Theni Medical College, Theni, Tamil Nadu, India.
| |
Collapse
|
28
|
Oyarzún P, Kobe B. Recombinant and epitope-based vaccines on the road to the market and implications for vaccine design and production. Hum Vaccin Immunother 2017; 12:763-7. [PMID: 26430814 DOI: 10.1080/21645515.2015.1094595] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Novel vaccination approaches based on rational design of B- and T-cell epitopes - epitope-based vaccines - are making progress in the clinical trial pipeline. The epitope-focused recombinant protein-based malaria vaccine (termed RTS,S) is a next-generation approach that successfully reached phase-III trials, and will potentially become the first commercial vaccine against a human parasitic disease. Progress made on methods such as recombinant DNA technology, advanced cell-culture techniques, immunoinformatics and rational design of immunogens are driving the development of these novel concepts. Synthetic recombinant proteins comprising both B- and T-cell epitopes can be efficiently produced through modern biotechnology and bioprocessing methods, and can enable the induction of large repertoires of immune specificities. In particular, the inclusion of appropriate CD4+ T-cell epitopes is increasingly considered a key vaccine component to elicit robust immune responses, as suggested by results coming from HIV-1 clinical trials. In silico strategies for vaccine design are under active development to address genetic variation in pathogens and several broadly protective "universal" influenza and HIV-1 vaccines are currently at different stages of clinical trials. Other methods focus on improving population coverage in target populations by rationally considering specificity and prevalence of the HLA proteins, though a proof-of-concept in humans has not been demonstrated yet. Overall, we expect immunoinformatics and bioprocessing methods to become a central part of the next-generation epitope-based vaccine development and production process.
Collapse
Affiliation(s)
- Patricio Oyarzún
- a Biotechnology Center, Facultad de Ingeniería y Tecnología, Universidad San Sebastián , Concepción , Chile
| | - Bostjan Kobe
- b School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Center, University of Queensland , Brisbane , Australia
| |
Collapse
|
29
|
Yu W, Wang L, Wang M, Liu S, Li W, Wang X, Li X, Yu S, Yao D, Ma J, Yu L, Chen J, Feng Z, Cui Y. Identification and characterization of CD4 + T cell epitopes on manganese transport protein C of Staphylococcus aureus. Microb Pathog 2017; 112:30-37. [PMID: 28942173 DOI: 10.1016/j.micpath.2017.09.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/26/2017] [Accepted: 09/19/2017] [Indexed: 11/29/2022]
Abstract
Manganese transport protein C (MntC) of Staphylococcus aureus represents an excellent vaccine-candidate antigen. The important role of CD4+ T cells in effective immunity against S. aureus infection was shown; however, CD4+ T cell-specific epitopes on S. aureus MntC have not been well identified. Here, we used bioinformatics prediction algorithms to evaluate and identify nine candidate epitopes within MntC. Our results showed that peptide M8 emulsified in Freund's adjuvant induced a much higher cell-proliferation rate as compared with controls. Additionally, CD4+ T cells stimulated with peptide M8 secreted significantly higher levels of interferon-γ and interleukin-17A. These results suggested that peptide M8 represented an H-2d (I-E)-restricted Th17-specific epitope.
Collapse
Affiliation(s)
- Wei Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Lizi Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Mengyao Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Shuo Liu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Wanyu Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Xintong Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Xiaoting Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Simiao Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Di Yao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Liquan Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Jing Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Zhenyue Feng
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China
| | - Yudong Cui
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, China.
| |
Collapse
|
30
|
Gu Y, Sun X, Li B, Huang J, Zhan B, Zhu X. Vaccination with a Paramyosin-Based Multi-Epitope Vaccine Elicits Significant Protective Immunity against Trichinella spiralis Infection in Mice. Front Microbiol 2017; 8:1475. [PMID: 28824599 PMCID: PMC5540943 DOI: 10.3389/fmicb.2017.01475] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
Trichinellosis is a worldwide zoonosis and remains a serious public health problem. Interrupting parasite transmission via vaccination of livestocks with a potent vaccine is a practical approach to prevent human Trichinellosis. Our previous studies have identified that paramyosin of Trichinella spiralis (Ts-Pmy) is a good vaccine candidate against Trichinellosis. In this study, a novel multi-epitope vaccine (MEP) was constructed by using four CD4+ T cell epitopes (P2, P3, P4, and P5) and one B cell epitope (YX1) from Ts-Pmy and expressed as a soluble recombinant protein (rMEP) in Escherichia coli. Mice immunized with rMEP vaccine produced significant higher muscle larval reduction (55.4%) than that induced by immunization of parental rTs-Pmy (34.4%) against T. spiralis infection. The better protection is associated with rMEP induced high levels of anti-rMEP specific IgG and subclass IgG1/IgG2a, elevated T cell proliferation of splenocytes and secretion of IFN-γ, IL-4 and IL-5. The cellular response to individual T cell epitope also showed that splenocytes from mice immunized with rMEP strongly response to the stimulation of synthetic epitope peptide P2, P3, and P4, but not to P5, suggesting that most of T cell epitopes are exposed and processed well during immunization that may contribute to the high protection induced by the immunization of rMEP. This study implies that epitope vaccine is a promising approach for the development of vaccines against Trichinellosis.
Collapse
Affiliation(s)
- Yuan Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| | - Ximeng Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| | - Bo Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| | - Bin Zhan
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, HoustonTX, United States
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| |
Collapse
|
31
|
Cunha-Neto E, Rosa DS, Harris PE, Olson T, Morrow A, Ciotlos S, Herst CV, Rubsamen RM. An Approach for a Synthetic CTL Vaccine Design against Zika Flavivirus Using Class I and Class II Epitopes Identified by Computer Modeling. Front Immunol 2017. [PMID: 28649242 PMCID: PMC5465239 DOI: 10.3389/fimmu.2017.00640] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The threat posed by severe congenital abnormalities related to Zika virus (ZKV) infection during pregnancy has turned development of a ZKV vaccine into an emergency. Recent work suggests that the cytotoxic T lymphocyte (CTL) response to infection is an important defense mechanism in response to ZKV. Here, we develop the rationale and strategy for a new approach to developing cytotoxic T lymphocyte (CTL) vaccines for ZKV flavivirus infection. The proposed approach is based on recent studies using a protein structure computer model for HIV epitope selection designed to select epitopes for CTL attack optimized for viruses that exhibit antigenic drift. Because naturally processed and presented human ZKV T cell epitopes have not yet been described, we identified predicted class I peptide sequences on ZKV matching previously identified DNV (Dengue) class I epitopes and by using a Major Histocompatibility Complex (MHC) binding prediction tool. A subset of those met the criteria for optimal CD8+ attack based on physical chemistry parameters determined by analysis of the ZKV protein structure encoded in open source Protein Data File (PDB) format files. We also identified candidate ZKV epitopes predicted to bind promiscuously to multiple HLA class II molecules that could provide help to the CTL responses. This work suggests that a CTL vaccine for ZKV may be possible even if ZKV exhibits significant antigenic drift. We have previously described a microsphere-based CTL vaccine platform capable of eliciting an immune response for class I epitopes in mice and are currently working toward in vivo testing of class I and class II epitope delivery directed against ZKV epitopes using the same microsphere-based vaccine.
Collapse
Affiliation(s)
- Edecio Cunha-Neto
- Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (III) INCT, São Paulo, Brazil.,School of Medicine, Heart Institute (Incor), University of São Paulo, São Paulo, Brazil
| | - Daniela S Rosa
- Institute for Investigation in Immunology (III) INCT, São Paulo, Brazil.,Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Paul E Harris
- Endocrinology Division, Department of Medicine, School of Medicine, Columbia University, New York, NY, United States
| | - Tim Olson
- Flow Pharma, Inc., Redwood City, CA, United States
| | - Alex Morrow
- Flow Pharma, Inc., Redwood City, CA, United States
| | | | | | - Reid Martin Rubsamen
- Flow Pharma, Inc., Redwood City, CA, United States.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
32
|
Li X, Guo L, Kong M, Su X, Yang D, Zou M, Liu Y, Lu L. Design and Evaluation of a Multi-Epitope Peptide of Human Metapneumovirus. Intervirology 2016; 58:403-12. [PMID: 27096202 DOI: 10.1159/000445059] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/23/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES No licensed vaccines or therapeutic agents for human metapneumovirus (hMPV) infection exist to date. We aimed to construct a multi-epitope peptide (MEP) of hMPV to show promising results for epitope-based vaccine development. METHODS Six independent algorithms were screened to predict B-cell epitopes of hMPV, and three algorithms were used to predict cytotoxic T lymphocyte and T helper (Th) lymphocyte epitopes. Predicted epitopes were assembled in series with the spacers GPGPG and KK introduced, termed MEP. Recombinant mep genes were inserted into pET32a(+) plasmid and expressed in Escherichia coli strain BL21 (DE3). BALB/c mice were immunized with MEP with different adjuvants. Antibody titer, lymphocyte proliferation, cytotoxic T lymphocyte (CTL) activity and splenocyte cytokines were detected 2 weeks later after the last immunization. Microneutralization assay was used to detect neutralizing antibodies. RESULTS Six B-cell epitopes, four CTL epitopes and two Th epitopes were screened to construct the mep gene. Expressed MEP induced >104 antibodies in BALB/c mice, and produced anti-MEP antibody reacting with hMPV strains specifically as detected in indirect fluorescent assay (the titer was 160). The lymphocyte proliferation index, CTL activity and splenocyte cytokines of the MEP immunization groups were higher than in the control group (p < 0.05). Both IgG1 and IgG2a antibodies could be detected in the different groups, and balanced Th1/Th2 cytokines were secreted by splenocytes in these groups. The mean neutralizing titers of the MEP+CpG ODN, MEP+Alum and MEP+Alum+ CpG ODN groups were 87 (95% CI 50-126), 93 (95% CI 67-121) and 96 (95% CI 69-147), respectively. CONCLUSION MEP of hMPV elicited both strong humoral immunity and cell-mediated immunity in mice. The anti-MEP serum could neutralize hMPV infection in vitro. Joint use of CpG ODN and aluminum hydroxide adjuvants obtained the best immune effects. This study may contribute to hMPV epitope-based vaccine development.
Collapse
Affiliation(s)
- Xiaoyan Li
- Tianjin Centers for Disease Control and Prevention, Tianjin, PR China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Okamoto M, Kobayashi M, Yonemitsu Y, Koido S, Homma S. Dendritic cell-based vaccine for pancreatic cancer in Japan. World J Gastrointest Pharmacol Ther 2016; 7:133-138. [PMID: 26855819 PMCID: PMC4734946 DOI: 10.4292/wjgpt.v7.i1.133] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/28/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023] Open
Abstract
“Vaccell” is a dendritic cell (DC)-based cancer vaccine which has been established in Japan. The DCs play central roles in deciding the direction of host immune reactions as well as antigen presentation. We have demonstrated that DCs treated with a streptococcal immune adjuvant OK-432, produce interleukin-12, induce Th1-dominant state, and elicit anti-tumor effects, more powerful than those treated with the known DC-maturating factors. We therefore decided to mature DCs by the OK-432 for making an effective DC vaccine, Vaccell. The 255 patients with inoperable pancreatic cancer who received standard chemotherapy combined with DC vaccines, were analyzed retrospectively. Survival time of the patients with positive delayed type hypersensitivity (DTH) skin reaction was significantly prolonged as compared with that of the patients with negative DTH. The findings strongly suggest that there may be “Responders” for the DC vaccine in advanced pancreatic cancer patients. We next conducted a small-scale prospective clinical study. In this trial, we pulsed HLA class II-restricted WT1 peptide (WT1-II) in addition to HLA class I-restricted peptide (WT1-I) into the DCs. Survival of the patients received WT1-I and -II pulsed DC vaccine was significantly extended as compared to that of the patients received DCs pulsed with WT1-I or WT1-II alone. Furthermore, WT1-specific DTH positive patients showed significantly improved the overall survival as well as progression-free survival as compared to the DTH negative patients. The activation of antigen-specific immune responses by DC vaccine in combination with standard chemotherapy may be associated with a good clinical outcome in advanced pancreatic cancer. We are now planning a pivotal study of the Vaccell in appropriate protocols in Japan.
Collapse
|
34
|
Rosa DS, Ribeiro SP, Fonseca SG, Almeida RR, Santana VC, Apostólico JDS, Kalil J, Cunha-Neto E. Multiple Approaches for Increasing the Immunogenicity of an Epitope-Based Anti-HIV Vaccine. AIDS Res Hum Retroviruses 2015; 31:1077-88. [PMID: 26149745 DOI: 10.1089/aid.2015.0101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The development of a highly effective vaccine against the human immunodeficiency virus (HIV) will likely be based on rational vaccine design, since traditional vaccine approaches have failed so far. In recent years, an understanding of what type of immune response is protective against infection and/or disease facilitated vaccine design. T cell-based vaccines against HIV have the goal of limiting both transmission and disease progression by inducing broad and functionally relevant T cell responses. In this context, CD4(+) T cells play a direct cytotoxic role and are also important for the generation and maintenance of functional CD8(+) T and B cell responses. The use of MHC-binding algorithms has allowed the identification of novel CD4(+) T cell epitopes that could be used in vaccine design, the so-called epitope-driven vaccine design. Epitope-based vaccines have the ability to focus the immune response on highly antigenic, conserved epitopes that are fully recognized by the target population. We have recently mapped a set of conserved multiple HLA-DR-binding HIV-1 CD4 epitopes and observed interferon (IFN)-γ-producing CD4(+) T cells when we tested these peptides in peripheral blood mononuclear cells (PBMCs) from HIV-infected individuals. We then designed multiepitopic DNA vaccines that induced broad and polyfunctional T cell responses in immunized mice. In this review we will focus on alternative strategies to increase the immunogenicity of an epitope-based vaccine against HIV infection.
Collapse
Affiliation(s)
- Daniela Santoro Rosa
- Departament of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Institute for Investigation in Immunology-INCT, São Paulo, Brazil
| | - Susan Pereira Ribeiro
- Institute for Investigation in Immunology-INCT, São Paulo, Brazil
- Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Rafael Ribeiro Almeida
- Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil
- Institute for Tropical Pathology and Public Health, Federal University of Goiás, Goiás, Brazil
| | - Vinicius Canato Santana
- Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil
- Institute for Tropical Pathology and Public Health, Federal University of Goiás, Goiás, Brazil
| | - Juliana de Souza Apostólico
- Departament of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Institute for Investigation in Immunology-INCT, São Paulo, Brazil
| | - Jorge Kalil
- Institute for Investigation in Immunology-INCT, São Paulo, Brazil
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Institute for Investigation in Immunology-INCT, São Paulo, Brazil
- Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
35
|
Oyarzun P, Kobe B. Computer-aided design of T-cell epitope-based vaccines: addressing population coverage. Int J Immunogenet 2015. [PMID: 26211755 DOI: 10.1111/iji.12214] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Epitope-based vaccines (EVs) make use of short antigen-derived peptides corresponding to immune epitopes, which are administered to trigger a protective humoral and/or cellular immune response. EVs potentially allow for precise control over the immune response activation by focusing on the most relevant - immunogenic and conserved - antigen regions. Experimental screening of large sets of peptides is time-consuming and costly; therefore, in silico methods that facilitate T-cell epitope mapping of protein antigens are paramount for EV development. The prediction of T-cell epitopes focuses on the peptide presentation process by proteins encoded by the major histocompatibility complex (MHC). Because different MHCs have different specificities and T-cell epitope repertoires, individuals are likely to respond to a different set of peptides from a given pathogen in genetically heterogeneous human populations. In addition, protective immune responses are only expected if T-cell epitopes are restricted by MHC proteins expressed at high frequencies in the target population. Therefore, without careful consideration of the specificity and prevalence of the MHC proteins, EVs could fail to adequately cover the target population. This article reviews state-of-the-art algorithms and computational tools to guide EV design through all the stages of the process: epitope prediction, epitope selection and vaccine assembly, while optimizing vaccine immunogenicity and coping with genetic variation in humans and pathogens.
Collapse
Affiliation(s)
- P Oyarzun
- Biotechnology Centre, Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Concepción, Chile
| | - B Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Oyarzun P, Ellis JJ, Gonzalez-Galarza FF, Jones AR, Middleton D, Boden M, Kobe B. A bioinformatics tool for epitope-based vaccine design that accounts for human ethnic diversity: application to emerging infectious diseases. Vaccine 2015; 33:1267-73. [PMID: 25629524 DOI: 10.1016/j.vaccine.2015.01.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/11/2014] [Accepted: 01/14/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Peptide vaccination based on multiple T-cell epitopes can be used to target well-defined ethnic populations. Because the response to T-cell epitopes is restricted by HLA proteins, the HLA specificity of T-cell epitopes becomes a major consideration for epitope-based vaccine design. We have previously shown that CD4+ T-cell epitopes restricted by 95% of human MHC class II proteins can be predicted with high-specificity. METHODS We describe here the integration of epitope prediction with population coverage and epitope selection algorithms. The population coverage assessment makes use of the Allele Frequency Net Database. We present the computational platform Predivac-2.0 for HLA class II-restricted epitope-based vaccine design, which accounts comprehensively for human genetic diversity. RESULTS We validated the performance of the tool on the identification of promiscuous and immunodominant CD4+ T-cell epitopes from the human immunodeficiency virus (HIV) protein Gag. We further describe an application for epitope-based vaccine design in the context of emerging infectious diseases associated with Lassa, Nipah and Hendra viruses. Putative CD4+ T-cell epitopes were mapped on the surface glycoproteins of these pathogens and are good candidates to be experimentally tested, as they hold potential to provide cognate help in vaccination settings in their respective target populations. CONCLUSION Predivac-2.0 is a novel approach in epitope-based vaccine design, particularly suited to be applied to virus-related emerging infectious diseases, because the geographic distributions of the viruses are well defined and ethnic populations in need of vaccination can be determined ("ethnicity-oriented approach"). Predivac-2.0 is accessible through the website http://predivac.biosci.uq.edu.au/.
Collapse
Affiliation(s)
- Patricio Oyarzun
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia; Biotechnology Centre, Universidad San Sebastián, Concepción, Chile.
| | - Jonathan J Ellis
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia
| | | | - Andrew R Jones
- Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Derek Middleton
- Transplant Immunology Laboratory, Royal Liverpool University Hospital & School of Infection and Host Defence University of Liverpool, United Kingdom
| | - Mikael Boden
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia; School of Information Technology and Electrical Engineering, University of Queensland, Queensland 4072, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Australia.
| |
Collapse
|
37
|
Felber BK, Valentin A, Rosati M, Bergamaschi C, Pavlakis GN. HIV DNA Vaccine: Stepwise Improvements Make a Difference. Vaccines (Basel) 2014; 2:354-79. [PMID: 26344623 PMCID: PMC4494255 DOI: 10.3390/vaccines2020354] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/11/2014] [Accepted: 04/18/2014] [Indexed: 12/15/2022] Open
Abstract
Inefficient DNA delivery methods and low expression of plasmid DNA have been major obstacles for the use of plasmid DNA as vaccine for HIV/AIDS. This review describes successful efforts to improve DNA vaccine methodology over the past ~30 years. DNA vaccination, either alone or in combination with other methods, has the potential to be a rapid, safe, and effective vaccine platform against AIDS. Recent clinical trials suggest the feasibility of its translation to the clinic.
Collapse
Affiliation(s)
- Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| |
Collapse
|
38
|
Abstract
The goal of active vaccination is to induce all the immune effector pathways and to establish immunological memory allowing prolonged surveillance against pathogens or cancer cells. DNA vaccination platform is an intriguing strategy owing to its ability to mobilize both branches of the immune system (i.e., innate immunity as well as adaptive immunity). Since plasmids offer several advantages for biotechnological applications due to their modular structure and easy manipulation, a wide range of strategies can be applied to improve DNA vaccine performance. This chapter discusses this topic in detail taking into account antigen/epitope selection and optimization, inclusion of intracellular targeting sequences and genetic adjuvants, and provision of T cell help.
Collapse
|
39
|
Abstract
A large volume of data relevant to immunology research has accumulated due to sequencing of genomes of the human and other model organisms. At the same time, huge amounts of clinical and epidemiologic data are being deposited in various scientific literature and clinical records. This accumulation of the information is like a goldmine for researchers looking for mechanisms of immune function and disease pathogenesis. Thus the need to handle this rapidly growing immunological resource has given rise to the field known as immunoinformatics. Immunoinformatics, otherwise known as computational immunology, is the interface between computer science and experimental immunology. It represents the use of computational methods and resources for the understanding of immunological information. It not only helps in dealing with huge amount of data but also plays a great role in defining new hypotheses related to immune responses. This chapter reviews classical immunology, different databases, and prediction tool. Further, it briefly describes applications of immunoinformatics in reverse vaccinology, immune system modeling, and cancer diagnosis and therapy. It also explores the idea of integrating immunoinformatics with systems biology for the development of personalized medicine. All these efforts save time and cost to a great extent.
Collapse
Affiliation(s)
- Namrata Tomar
- Machine Intelligence Unit, Indian Statistical Institute, 203 B.T. Road, Kolkata, 700108, India,
| | | |
Collapse
|
40
|
Cole GT, Hung CY, Sanderson SD, Hurtgen BJ, Wüthrich M, Klein BS, Deepe GS, Ostroff GR, Levitz SM. Novel strategies to enhance vaccine immunity against coccidioidomycosis. PLoS Pathog 2013; 9:e1003768. [PMID: 24367252 PMCID: PMC3868515 DOI: 10.1371/journal.ppat.1003768] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Garry T. Cole
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| | - Chiung-Yu Hung
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Sam D. Sanderson
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Brady J. Hurtgen
- U.S. Army Institute of Surgical Research, Fort Sam Houston, San Antonio, Texas, United States of America
| | - Marcel Wüthrich
- Department of Pediatrics and Microbiology and Immunology, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Bruce S. Klein
- Department of Pediatrics and Microbiology and Immunology, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - George S. Deepe
- Division of Infectious Diseases, College of Medicine, University of Cincinnati and Veterans Affairs Hospital, Cincinnati, Ohio, United States of America
| | - Gary R. Ostroff
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Stuart M. Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
41
|
Gu Y, Wei J, Yang J, Huang J, Yang X, Zhu X. Protective immunity against Trichinella spiralis infection induced by a multi-epitope vaccine in a murine model. PLoS One 2013; 8:e77238. [PMID: 24130862 PMCID: PMC3795051 DOI: 10.1371/journal.pone.0077238] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 08/30/2013] [Indexed: 12/13/2022] Open
Abstract
Trichinellosis is one of the most important food-borne parasitic zoonoses throughout the world. Because infected pigs are the major source of human infections, and China is becoming the largest international producer of pork, the development of a transmission-blocking vaccine to prevent swine from being infected is urgently needed for trichinellosis control in China. Our previous studies have demonstrated that specific Trichinella spiralis paramyosin (Ts-Pmy) and Ts-87 antigen could provide protective immunity against T. spiralis infection in immunized mice. Certain protective epitopes of Ts-Pmy and Ts-87 antigen have been identified. To identify more Ts-Pmy protective epitopes, a new monoclonal antibody, termed 8F12, was produced against the N-terminus of Ts-Pmy. This antibody elicited significant protective immunity in mice against T. spiralis infection by passive transfer and was subsequently used to screen a random phage display peptide library to identify recognized epitopes. Seven distinct positive phage clones were identified and their displayed peptides were sequenced. Synthesized epitope peptides conjugated to keyhole limpet hemocyanin were used to immunize mice, four of which exhibited larval reduction (from 18.7% to 26.3%, respectively) in vaccinated mice in comparison to the KLH control. To increase more effective protection, the epitope 8F7 that was found to induce the highest protection in this study was combined with two other previously identified epitopes (YX1 from Ts-Pmy and M7 from Ts-87) to formulate a multi-epitope vaccine. Mice immunized with this multi-epitope vaccine experienced a 35.0% reduction in muscle larvae burden after being challenged with T. spiralis larvae. This protection is significantly higher than that induced by individual-epitope peptides and is associated with high levels of subclasses IgG and IgG1. These results showed that a multi-epitope vaccine induced better protective immunity than an individual epitope and provided a feasible approach for developing a safer and more effective vaccine against trichinellosis.
Collapse
Affiliation(s)
- Yuan Gu
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Junfei Wei
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Jing Yang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Jingjing Huang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Xiaodi Yang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Xinping Zhu
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
- * E-mail:
| |
Collapse
|
42
|
Grubaugh D, Flechtner JB, Higgins DE. Proteins as T cell antigens: Methods for high-throughput identification. Vaccine 2013; 31:3805-10. [DOI: 10.1016/j.vaccine.2013.06.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/03/2013] [Accepted: 06/13/2013] [Indexed: 12/22/2022]
|
43
|
NOYA VERÓNICA, BAY SYLVIE, FESTARI MARÍAFLORENCIA, GARCÍA ENRIQUEP, RODRIGUEZ ERNESTO, CHIALE CAROLINA, GANNEAU CHRISTELLE, BALEUX FRANÇOISE, ASTRADA SOLEDAD, BOLLATI-FOGOLÍN MARIELA, OSINAGA EDUARDO, FREIRE TERESA. Mucin-like peptides from Echinococcus granulosus induce antitumor activity. Int J Oncol 2013; 43:775-84. [DOI: 10.3892/ijo.2013.2000] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/30/2013] [Indexed: 11/06/2022] Open
|
44
|
Oyarzún P, Ellis JJ, Bodén M, Kobe B. PREDIVAC: CD4+ T-cell epitope prediction for vaccine design that covers 95% of HLA class II DR protein diversity. BMC Bioinformatics 2013; 14:52. [PMID: 23409948 PMCID: PMC3598884 DOI: 10.1186/1471-2105-14-52] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/31/2013] [Indexed: 12/18/2022] Open
Abstract
Background CD4+ T-cell epitopes play a crucial role in eliciting vigorous protective immune responses during peptide (epitope)-based vaccination. The prediction of these epitopes focuses on the peptide binding process by MHC class II proteins. The ability to account for MHC class II polymorphism is critical for epitope-based vaccine design tools, as different allelic variants can have different peptide repertoires. In addition, the specificity of CD4+ T-cells is often directed to a very limited set of immunodominant peptides in pathogen proteins. The ability to predict what epitopes are most likely to dominate an immune response remains a challenge. Results We developed the computational tool Predivac to predict CD4+ T-cell epitopes. Predivac can make predictions for 95% of all MHC class II protein variants (allotypes), a substantial advance over other available methods. Predivac bases its prediction on the concept of specificity-determining residues. The performance of the method was assessed both for high-affinity HLA class II peptide binding and CD4+ T-cell epitope prediction. In terms of epitope prediction, Predivac outperformed three available pan-specific approaches (delivering the highest specificity). A central finding was the high accuracy delivered by the method in the identification of immunodominant and promiscuous CD4+ T-cell epitopes, which play an essential role in epitope-based vaccine design. Conclusions The comprehensive HLA class II allele coverage along with the high specificity in identifying immunodominant CD4+ T-cell epitopes makes Predivac a valuable tool to aid epitope-based vaccine design in the context of a genetically heterogeneous human population.The tool is available at: http://predivac.biosci.uq.edu.au/.
Collapse
Affiliation(s)
- Patricio Oyarzún
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | |
Collapse
|
45
|
Cole GT, Hurtgen BJ, Hung CY. Progress Toward a Human Vaccine Against Coccidioidomycosis. CURRENT FUNGAL INFECTION REPORTS 2012; 6:235-244. [PMID: 23585916 DOI: 10.1007/s12281-012-0105-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Coccidioidomycosis (San Joaquin Valley fever) is a human respiratory disease caused by a soil-borne mold, and is recognized as an intransigent microbial infection by physicians who treat patients with the potentially life-threatening, disseminated form of this mycosis. Epidemiological studies based on surveys of skin-test reactivity of people who reside in the endemic regions of the Southwestern US have shown that at least 150,000 new infections occur annually. The clinical spectrum of coccidioidomycosis ranges from an asymptomatic insult to a severe pulmonary disease in which the pathogen may spread from the lungs to the skin, bones, brain and other body organs. Escalation of symptomatic infections and increased cost of long-term antifungal treatment warrant a concerted effort to develop a vaccine against coccidioidomycosis. This review examines recently reported strategies used to generate such a vaccine and summarizes current understanding of the nature of protective immunity to this formidable disease.
Collapse
Affiliation(s)
- Garry T Cole
- Department of Biology and South Texas Center for Emerging Infectious Diseases (STCEID), University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | | | | |
Collapse
|
46
|
Construction and evaluation of a novel recombinant T cell epitope-based vaccine against Coccidioidomycosis. Infect Immun 2012; 80:3960-74. [PMID: 22949556 DOI: 10.1128/iai.00566-12] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Clinical and animal studies of coccidioidomycosis have demonstrated that activated CD4(+) T lymphocytes are essential for protection against this fungal respiratory disease. We previously reported a vaccine against Coccidioides infection which contained three recombinant CD4(+) T cell-reactive proteins and induced a robust, protective immune response in mice. Due to the anticipated high cost of production and clinical assessment of this multivalent vaccine, we generated a single protein which contained immunodominant T cell epitopes of the three polypeptides. Epitopes were initially identified by computational prediction of their ability to bind promiscuously to human major histocompatibility complex class II (MHC II) molecules. Cellular immunoassays confirmed the immunogenicity of the synthesized epitope peptides, while in vitro binding assays revealed a range of peptide affinity for MHC II. A DNA construct was synthesized for bacterial expression of a recombinant protein vaccine which contained five epitopes with the highest affinity for human MHC II, each fused with leader and spacer peptides proposed to optimize epitope processing and presentation to T cell receptors. Recall assays of immune T lymphocytes obtained from human MHC II-expressing HLA-DR4 transgenic mice confirmed that 4 of the 5 epitope peptides were processed. Mice immunized with the epitope-based vaccine admixed with a synthetic oligodeoxynucleotide adjuvant or loaded into yeast glucan particles and then challenged intranasally with Coccidioides showed early lung infiltration of activated T helper-1 (Th1), Th2, and Th17 cells, elevated gamma interferon (IFN-γ) and interleukin (IL)-17 production, significant reduction of fungal burden, and prolongation of survival compared to nonvaccinated mice. This is the first report of an epitope-based vaccine against coccidioidomycosis.
Collapse
|
47
|
Epitope based recombinant BCG vaccine elicits specific Th1 polarized immune responses in BALB/c mice. Vaccine 2012; 30:1364-70. [DOI: 10.1016/j.vaccine.2011.12.059] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 12/05/2011] [Accepted: 12/12/2011] [Indexed: 11/24/2022]
|
48
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
49
|
Iurescia S, Fioretti D, Fazio VM, Rinaldi M. Epitope-driven DNA vaccine design employing immunoinformatics against B-cell lymphoma: A biotech's challenge. Biotechnol Adv 2012; 30:372-83. [DOI: 10.1016/j.biotechadv.2011.06.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 06/16/2011] [Accepted: 06/23/2011] [Indexed: 12/16/2022]
|
50
|
Bonvalet M, Wambre E, Moussu H, Horiot S, Kwok WW, Louise A, Ebo D, Hoarau C, Van Overtvelt L, Baron-Bodo V, Moingeon P. Comparison between major histocompatibility complex class II tetramer staining and surface expression of activation markers for the detection of allergen-specific CD4⁺ T cells. Clin Exp Allergy 2011; 41:821-9. [PMID: 21418343 DOI: 10.1111/j.1365-2222.2011.03708.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Major histocompatibility complex (MHC) class II tetramers (tetramers) allow to detect allergen-specific CD4(+) T cells at a single-cell level. Limits to this technology include HLA restriction and the need to identify immunodominant T cell epitopes. OBJECTIVE Assessing the expression of various activation markers following allergen stimulation to replace tetramer staining. METHODS Peripheral blood mononuclear cells (PBMCs) from 25 birch pollen, grass pollen or house dust mite allergic individuals were stimulated with peptide mixes encompassing immunodominant epitopes from corresponding major allergens. After 2 weeks of in vitro amplification, cells were stained with both the appropriate tetramer and antibodies directed to CD25, CD30, CD39, CD69, CD137, CD154, GITR, HLA-DR and ICOS, before FACS analysis. RESULTS Following allergen stimulation, percentages of tetramer(+) cells among CD4(+) CD154(+) cells range from 5% to 87%, depending upon donors. As for CD154, a large inter-individual variability is observed in terms of surface expression for all activation markers tested in allergen-stimulated PBMCs. T cells reactive with either tetramers (0.4-10.4% CD4(+) T cells) or anti-marker antibodies (2.2-32.7% CD4(+) T cells), but not both, are observed, reflecting the presence of anergic as well as non-specifically activated cells. Tetramer(+) /marker(+) , tetramer(+) /marker(-) and tetramer(-) /marker(+) cells were compared for their capacity to express cytokines, demonstrating that only the former represent bona fide allergen-specific activated CD4(+) T cells, based upon a higher expression of cytokines or corresponding genes in presence of the allergen. CONCLUSION AND CLINICAL RELEVANCE No strict correlation exists between tetramer staining and the expression of multiple activation markers in stimulated CD4(+) T cells. Dual staining allows to discriminate functional tetramer(+) /marker(+) vs. anergic (tetramer(+) /marker(-) ) allergen-specific T cells or non-specifically activated (tetramer(-) /marker(+) ) T cells. Combining tetramer staining with the detection of activation markers helps understanding patient heterogeneity regarding specific CD4(+) T cell responses. This approach has immediate relevance for monitoring immune changes induced during specific immunotherapy.
Collapse
|