1
|
Kanwal M, Basheer A, Bilal M, Faheem M, Aziz T, Alamri AS, Alsanie WF, Alhomrani M, Jamal SB. In silico vaccine design for Yersinia enterocolitica: A comprehensive approach to enhanced immunogenicity, efficacy and protection. Int Immunopharmacol 2024; 143:113241. [PMID: 39369465 DOI: 10.1016/j.intimp.2024.113241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Yersinia enterocolitica, a foodborne pathogen, has emerged as a significant public health concern due to its increased prevalence and multidrug resistance. This study employed reverse vaccinology to identify novel vaccine candidates against Y. enterocolitica through comprehensive in silico analyses. The core genome's conserved protein translocase subunit SecY was selected as the target, and potential B-cell, MHC class I, and MHC class II epitopes were mapped. 3B-cell epitopes, 3 MHCI and 11 MHCII epitopes were acquired. A multi-epitope vaccine construct was designed by incorporating the identified epitopes, TLR4 Agonist was used as adjuvants to enhance the immunogenic response. EAAAK, CPGPG and AYY linkers were used to form a vaccine construct, followed by extensive computational evaluations. The vaccine exhibited desirable physicochemical properties, stable secondary and tertiary structures as evaluated by PDBSum and trRosetta. Moreover, favorable interactions with the human Toll-like receptor 4 (TLR4) was observed by ClusPro. Population coverage analysis estimated the vaccine's applicability across 99.74 % in diverse populations. In addition, molecular dynamics simulations and normal mode analysis confirmed the vaccine's structural stability and dynamics in a simulated biological environment. Furthermore, codon optimization and in silico cloning facilitated the evaluation of the vaccine's expression potential in E. coli and pET-28a was used a recombinant plasmid. This study provides a promising foundation for the development of an efficacious vaccine against Y. enterocolitica infections.
Collapse
Affiliation(s)
- Munazza Kanwal
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| | - Amina Basheer
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| | - Muhammad Bilal
- Department of Biological Sciences, Oakland University, MI, USA.
| | - Muhammad Faheem
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202, USA.
| | - Tariq Aziz
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece.
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia.
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia.
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia.
| | - Syed Babar Jamal
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| |
Collapse
|
2
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
3
|
Liu Y, Zhou J, Yang Y, Chen X, Chen L, Wu Y. Intestinal Microbiota and Its Effect on Vaccine-Induced Immune Amplification and Tolerance. Vaccines (Basel) 2024; 12:868. [PMID: 39203994 PMCID: PMC11359036 DOI: 10.3390/vaccines12080868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
This review provides the potential of intestinal microbiota in vaccine design and application, exploring the current insights into the interplay between the intestinal microbiota and the immune system, with a focus on its intermediary function in vaccine efficacy. It summarizes families and genera of bacteria that are part of the intestinal microbiota that may enhance or diminish vaccine efficacy and discusses the foundational principles of vaccine sequence design and the application of gut microbial characteristics in vaccine development. Future research should further investigate the use of multi-omics technologies to elucidate the interactive mechanisms between intestinal microbiota and vaccine-induced immune responses, aiming to optimize and improve vaccine design.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Xiangzheng Chen
- Department of Liver Surgery & Liver Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Yangping Wu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
| |
Collapse
|
4
|
Lim CP, Leow CH, Lim HT, Kok BH, Chuah C, Oliveira JIN, Jones M, Leow CY. Insights into structural vaccinology harnessed for universal coronavirus vaccine development. Clin Exp Vaccine Res 2024; 13:202-217. [PMID: 39144127 PMCID: PMC11319108 DOI: 10.7774/cevr.2024.13.3.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 08/16/2024] Open
Abstract
Structural vaccinology is pivotal in expediting vaccine design through high-throughput screening of immunogenic antigens. Leveraging the structural and functional characteristics of antigens and immune cell receptors, this approach employs protein structural comparison to identify conserved patterns in key pathogenic components. Molecular modeling techniques, including homology modeling and molecular docking, analyze specific three-dimensional (3D) structures and protein interactions and offer valuable insights into the 3D interactions and binding affinity between vaccine candidates and target proteins. In this review, we delve into the utilization of various immunoinformatics and molecular modeling tools to streamline the development of broad-protective vaccines against coronavirus disease 2019 variants. Structural vaccinology significantly enhances our understanding of molecular interactions between hosts and pathogens. By accelerating the pace of developing effective and targeted vaccines, particularly against the rapidly mutating severe acute respiratory syndrome coronavirus 2 and other prevalent infectious diseases, this approach stands at the forefront of advancing immunization strategies. The combination of computational techniques and structural insights not only facilitates the identification of potential vaccine candidates but also contributes to the rational design of vaccines, fostering a more efficient and targeted approach to combatting infectious diseases.
Collapse
Affiliation(s)
- Chin Peng Lim
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Hui Ting Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Boon Hui Kok
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Candy Chuah
- Faculty of Medicine, Asian Institute of Medical Science and Technology University, Bedong, Malaysia
| | - Jonas Ivan Nobre Oliveira
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Malcolm Jones
- School of Veterinary Science, The University of Queensland, Gatton, Australia
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
| |
Collapse
|
5
|
Bravi B. Development and use of machine learning algorithms in vaccine target selection. NPJ Vaccines 2024; 9:15. [PMID: 38242890 PMCID: PMC10798987 DOI: 10.1038/s41541-023-00795-8] [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: 08/04/2023] [Accepted: 12/07/2023] [Indexed: 01/21/2024] Open
Abstract
Computer-aided discovery of vaccine targets has become a cornerstone of rational vaccine design. In this article, I discuss how Machine Learning (ML) can inform and guide key computational steps in rational vaccine design concerned with the identification of B and T cell epitopes and correlates of protection. I provide examples of ML models, as well as types of data and predictions for which they are built. I argue that interpretable ML has the potential to improve the identification of immunogens also as a tool for scientific discovery, by helping elucidate the molecular processes underlying vaccine-induced immune responses. I outline the limitations and challenges in terms of data availability and method development that need to be addressed to bridge the gap between advances in ML predictions and their translational application to vaccine design.
Collapse
Affiliation(s)
- Barbara Bravi
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
6
|
Bahramali G, Mashhadi Abolghasem Shirazi M, Hannan M, Aghasadeghi MR, Khosravy MS, Arjmand S, Sadat SM. Immunogenicity evaluation of a novel virus-like particle vaccine candidate against SARS-CoV-2 in BALB/c. Pathog Dis 2023; 81:ftad021. [PMID: 37667486 DOI: 10.1093/femspd/ftad021] [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: 07/19/2023] [Revised: 08/12/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
The coronavirus disease (COVID-19) pandemic has imposed deployment of an effective vaccine as a worldwide health priority. The new variants of SARS-CoV-2 have also brought serious concerns due to virus eradiation hesitancy. In this study, we evaluated the protective immune system activity of a recombinant viral vector-based vaccine candidate encoding a fusion spike, membrane and nucleocapsid proteins, Spike (528-1273aa)-M-N, in BALB/c via two different routes of delivery, intranasal and subcutaneous. The immune responses were then assessed through specific SARS-CoV-2 antibodies, interleukin and granzyme B secretion. The outcomes showed that the IgG titer and IgA secretion was higher in intranasal route in comparison with the subcutaneous, and what is more, a higher titer of IL-4 was detected through the intranasal route, whereas IFN-γ was highly induced via the subcutaneous route. The cytotoxic cell activities were mostly achieved via subcutaneous route immunization. Vaccination with the target antigen is immunogenic and led to induction of specific antibodies. Both humoral and cellular immunity arms were well activated in immunized mice, especially through intranasal route with detectable IgA and IgG. Therefore, implication of the platform as a potential vaccine candidate has potential as a future prophylactic vaccine that guarantees further investigations for the assessment of its immunogenicity in humans.
Collapse
Affiliation(s)
- Golnaz Bahramali
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | | | - Mina Hannan
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Mohammad Reza Aghasadeghi
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Mohammad Sadeq Khosravy
- Department of Rabies Research (B), WHO Collaborating Center for Reference and Research on Rabies, Tehran 1316946551, Iran
| | - Sina Arjmand
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| |
Collapse
|
7
|
Capelli R, Serapian SA, Colombo G. Computational Epitope Prediction and Design for Antibody Development and Detection. Methods Mol Biol 2023; 2552:255-266. [PMID: 36346596 DOI: 10.1007/978-1-0716-2609-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The design of optimized protein antigens is a fundamental step in the development of new vaccine candidates and in the detection of therapeutic antibodies. A fundamental prerequisite is the identification of antigenic regions that are most prone to interact with antibodies, namely, B-cell epitopes. Here, we describe an efficient structure-based computational method for epitope prediction, called MLCE. In this approach, all that is required is the 3D structure of the antigen of interest. MLCE can be applied to glycosylated proteins, facilitating the identification of immunoreactive versus immune-shielding carbohydrates.
Collapse
Affiliation(s)
- Riccardo Capelli
- SCITEC-CNR, Milan, Italy
- Politecnico di Torino, Department of Applied Science and Technology, Torino, Italy
| | | | - Giorgio Colombo
- SCITEC-CNR, Milan, Italy.
- Università di Pavia, Dipartimento di Chimica, Pavia, Italy.
| |
Collapse
|
8
|
Kawamura H, Yoshino N, Murakami K, Kawamura H, Sugiyama I, Sasaki Y, Odagiri T, Sadzuka Y, Muraki Y. The relationship between the chemical structure, physicochemical properties, and mucosal adjuvanticity of sugar-based surfactants. Eur J Pharm Biopharm 2023; 182:1-11. [PMID: 36455784 DOI: 10.1016/j.ejpb.2022.11.023] [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: 08/29/2022] [Revised: 11/07/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The relationship between the chemical structure, physicochemical properties, and mucosal adjuvanticity of sugar-based surfactants (SBSs) has not been sufficiently elucidated. Thus, in the present study, we systematically analyzed 11 SBSs for mucosal adjuvanticity. Ovalbumin (OVA)-specific antibody titers were measured in mice immunized intranasally with OVA plus SBS. We found that four SBSs (trehalose monododecanoate, sucrose monododecanoate, n-dodecyl-α-d-maltopyranoside, and n-dodecyl-β-d-maltopyranoside) exhibited the most potent adjuvanticity. We identified the following associations between chemical structure and adjuvanticity: 1) OVA-specific antibody titer increased with an increasing number of carbon atoms in the alkyl chain; 2) the adjuvanticity was not affected by the type of sugar or bond between the sugar and alkyl chain; and 3) SBSs with rigid structures exhibited less adjuvanticity. The relationship between physicochemical properties and adjuvanticity was as follows: 1) SBSs exhibited adjuvanticity above the critical micelle concentration and 2) in the SBSs with potent adjuvanticity, the diameter of the SBS-OVA complex was 70-75 nm. Our study indicates evidence for the direct involvement of chemical structure and physicochemical properties in determining adjuvanticity in SBSs.
Collapse
Affiliation(s)
- Hanae Kawamura
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Naoto Yoshino
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan.
| | - Kazuyuki Murakami
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Hideki Kawamura
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Ikumi Sugiyama
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Sciences, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yutaka Sasaki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Takashi Odagiri
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yasuyuki Sadzuka
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Sciences, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yasushi Muraki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| |
Collapse
|
9
|
Khan MA, Amin A, Farid A, Ullah A, Waris A, Shinwari K, Hussain Y, Alsharif KF, Alzahrani KJ, Khan H. Recent Advances in Genomics-Based Approaches for the Development of Intracellular Bacterial Pathogen Vaccines. Pharmaceutics 2022; 15:pharmaceutics15010152. [PMID: 36678781 PMCID: PMC9863128 DOI: 10.3390/pharmaceutics15010152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Infectious diseases continue to be a leading cause of morbidity and mortality worldwide. The majority of infectious diseases are caused by intracellular pathogenic bacteria (IPB). Historically, conventional vaccination drives have helped control the pathogenesis of intracellular bacteria and the emergence of antimicrobial resistance, saving millions of lives. However, in light of various limitations, many diseases that involve IPB still do not have adequate vaccines. In response to increasing demand for novel vaccine development strategies, a new area of vaccine research emerged following the advent of genomics technology, which changed the paradigm of vaccine development by utilizing the complete genomic data of microorganisms against them. It became possible to identify genes related to disease virulence, genetic patterns linked to disease virulence, as well as the genetic components that supported immunity and favorable vaccine responses. Complete genomic databases, and advancements in transcriptomics, metabolomics, structural genomics, proteomics, immunomics, pan-genomics, synthetic genomics, and population biology have allowed researchers to identify potential vaccine candidates and predict their effects in patients. New vaccines have been created against diseases for which previously there were no vaccines available, and existing vaccines have been improved. This review highlights the key issues and explores the evolution of vaccines. The increasing volume of IPB genomic data, and their application in novel genome-based techniques for vaccine development, were also examined, along with their characteristics, and the opportunities and obstacles involved. Critically, the application of genomics technology has helped researchers rapidly select and evaluate candidate antigens. Novel vaccines capable of addressing the limitations associated with conventional vaccines have been developed and pressing healthcare issues are being addressed.
Collapse
Affiliation(s)
- Muhammad Ajmal Khan
- Division of Life Science, Center for Cancer Research, and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
- Correspondence: (M.A.K.); or (H.K.)
| | - Aftab Amin
- Division of Life Science, Center for Cancer Research, and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Awais Farid
- Division of Environment and Sustainability, Hong Kong University of Science and Technology, Hong Kong, China
| | - Amin Ullah
- Molecular Virology Laboratory, Department of Microbiology and Biotechnology, Abasyn University, Peshawar 25000, Pakistan
| | - Abdul Waris
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Khyber Shinwari
- Institute of Chemical Engineering, Department Immuno-Chemistry, Ural Federal University, Yekaterinbiurg 620002, Russia
| | - Yaseen Hussain
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Khalid J. Alzahrani
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Haroon Khan
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Correspondence: (M.A.K.); or (H.K.)
| |
Collapse
|
10
|
Kumar V, Kumar S, Sharma PC. Recent advances in the vaccine development for the prophylaxis of SARS Covid-19. Int Immunopharmacol 2022; 111:109175. [PMID: 35994853 PMCID: PMC9381430 DOI: 10.1016/j.intimp.2022.109175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused Coronavirus Disease 2019 (COVID-19) is currently a global pandemic that has wreaked havoc on public health, lives, and the global economy. The present COVID-19 outbreak has put pressure on the scientific community to develop medications and vaccinations to combat COVID-19. However, according to highly optimistic forecasts, we could not have a COVID-19 vaccine until September 2020. This is due to the fact that a successful COVID-19 vaccine will necessitate a careful validation of effectiveness and adverse reactivity given that the target vaccine population includes high-risk people over 60, particularly those with severe co-morbid conditions, frontline healthcare professionals, and those involved in essential industrial sectors. For passive immunization, which is being considered for Covid-19, there are several platforms for vaccine development, each with its own advantages and disadvantages. The COVID-19 pandemic, which is arguably the deadliest in the last 100 years after the Spanish flu, necessitates a swift assessment of the various approaches for their ability to incite protective immunity and safety to prevent unintended immune potentiation, which is crucial to the pathogenesis of this virus. Considering the pandemic's high fatality rate and rapid spread, an efficient vaccination is critical for its management. As a result, academia, industry, and government are collaborating in unprecedented ways to create and test a wide range of vaccinations. In this review, we summarize the Covid-19 vaccine development initiatives, recent trends, difficulties, comparison between traditional vaccines development and Covid-19 vaccines development also listed the approved/authorized, phase-3 and pre-clinical trials Covid-19 vaccines in different countries.
Collapse
Affiliation(s)
- Vipul Kumar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Sahil Kumar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India.
| | - Prabodh Chander Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| |
Collapse
|
11
|
Han X, Cai Z, Dai Y, Huang H, Cao X, Wang Y, Fang Y, Liu G, Zhang M, Zhang Y, Yang B, Xue W, Zhao G, Tai W, Li M. Re-burying Artificially Exposed Surface of Viral Subunit Vaccines Through Oligomerization Enhances Vaccine Efficacy. Front Cell Infect Microbiol 2022; 12:927674. [PMID: 35846760 PMCID: PMC9278648 DOI: 10.3389/fcimb.2022.927674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/02/2022] [Indexed: 12/02/2022] Open
Abstract
Viral subunit vaccines often suffer low efficacy. We recently showed that when taken out of the context of whole virus particles, recombinant subunit vaccines contain artificially exposed surface regions that are non-neutralizing and reduce their efficacy, and thus these regions need to be re-buried in vaccine design. Here we used the envelope protein domain III (EDIII) of Japanese encephalitis virus (JEV), a subunit vaccine candidate, to further validate this important concept for subunit vaccine designs. We constructed monomeric EDIII, dimeric EDIII via a linear space, dimeric EDIII via an Fc tag, and trimeric EDIII via a foldon tag. Compared to monomeric EDIII or linearly linked dimeric EDIII, tightly packed EDIII oligomers via the Fc or foldon tag induce higher neutralizing antibody titers in mice and also protect mice more effectively from lethal JEV challenge. Structural analyses demonstrate that part of the artificially exposed surface areas on recombinant EDIII becomes re-buried in Fc or foldon-mediated oligomers. This study further establishes the artificially exposed surfaces as an intrinsic limitation of subunit vaccines, and suggests that re-burying these surfaces through tightly packed oligomerization is a convenient and effective approach to overcome this limitation.
Collapse
Affiliation(s)
- Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhuming Cai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yulong Dai
- Institute of Hemu Biotechnology, Beijing Hemu Biotechnology Co., Ltd, Beijing, China
| | - He Huang
- Institute of Hemu Biotechnology, Beijing Hemu Biotechnology Co., Ltd, Beijing, China
| | - Xiangwen Cao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.,Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yingying Fang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Gang Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Min Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yuhang Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Binhui Yang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Wei Xue
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.,Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Alves MLF, Ferreira MRA, Rodrigues RR, Conceição FR. Clostridium haemolyticum, a review of beta toxin and insights into the antigen design for vaccine development. Mol Immunol 2022; 148:45-53. [PMID: 35665660 DOI: 10.1016/j.molimm.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/01/2022] [Accepted: 05/16/2022] [Indexed: 01/03/2023]
Abstract
Phospholipases C (PLCs) represent an important group of lethal toxins produced by pathogenic bacteria of the Clostridium genus, including the beta toxin of C. haemolyticum. Bacillary hemoglobinuria in cattle and sheep is the main disease caused by this pathogen and its incidence can be reduced by annual vaccination of herds. Currently, widely used vaccines depend on cultivating the pathogen and obtaining high concentrations of the toxin, disadvantages that can be overcome with the use of recombinant vaccines. In the development of this new generation of immunizing agents, identifying and understanding the structural and immunological aspects of the antigen are crucial steps, but despite this, the beta toxin is poorly characterized. Fortunately, the time and resources required for these investigations can be reduced using immunoinformatics. To advance the development of recombinant vaccines, in addition to a brief review of the structural and immunological aspects of beta toxin, this work provides in silico mapping of immunodominant regions to guide future vaccinology studies against C. haemolyticum. A review of alternatives to overcome the limitations of beta toxin vaccines (conventional or recombinant) is also proposed.
Collapse
Affiliation(s)
- Mariliana Luiza Ferreira Alves
- Instituto Federal Sul-rio-grandense - IFSUL, Praça Vinte de Setembro, 455, Centro, Pelotas CEP 96.015-360, RS, Brazil; Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil.
| | - Marcos Roberto Alves Ferreira
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil
| | - Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil
| |
Collapse
|
13
|
Vishweshwaraiah YL, Dokholyan NV. Toward rational vaccine engineering. Adv Drug Deliv Rev 2022; 183:114142. [PMID: 35150769 PMCID: PMC8931536 DOI: 10.1016/j.addr.2022.114142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/29/2022]
Abstract
Technological revolutions in several fields have pushed the boundaries of vaccine design and provided new avenues for vaccine development. Next-generation vaccine platforms have shown promise in targeting challenging antigens, for which traditional approaches have been ineffective. With advances in protein engineering, structural biology, computational biology and immunology, the structural vaccinology approach, which uses protein structure information to develop immunogens, holds promise for future vaccine design. In this review, we highlight various vaccine development strategies, along with their advantages and limitations. We discuss the rational vaccine design approach, which focuses on structure-based vaccine design. Finally, we discuss antigen engineering using the epitope-scaffold approach, gaps in structural vaccinology, and remaining challenges in vaccine design.
Collapse
Affiliation(s)
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033-0850, USA; Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033-0850, USA; Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
14
|
Amimo JO, Raev SA, Chepngeno J, Mainga AO, Guo Y, Saif L, Vlasova AN. Rotavirus Interactions With Host Intestinal Epithelial Cells. Front Immunol 2021; 12:793841. [PMID: 35003114 PMCID: PMC8727603 DOI: 10.3389/fimmu.2021.793841] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
Rotavirus (RV) is the foremost enteric pathogen associated with severe diarrheal illness in young children (<5years) and animals worldwide. RV primarily infects mature enterocytes in the intestinal epithelium causing villus atrophy, enhanced epithelial cell turnover and apoptosis. Intestinal epithelial cells (IECs) being the first physical barrier against RV infection employs a range of innate immune strategies to counteract RVs invasion, including mucus production, toll-like receptor signaling and cytokine/chemokine production. Conversely, RVs have evolved numerous mechanisms to escape/subvert host immunity, seizing translation machinery of the host for effective replication and transmission. RV cell entry process involve penetration through the outer mucus layer, interaction with cell surface molecules and intestinal microbiota before reaching the IECs. For successful cell attachment and entry, RVs use sialic acid, histo-blood group antigens, heat shock cognate protein 70 and cell-surface integrins as attachment factors and/or (co)-receptors. In this review, a comprehensive summary of the existing knowledge of mechanisms underlying RV-IECs interactions, including the role of gut microbiota, during RV infection is presented. Understanding these mechanisms is imperative for developing efficacious strategies to control RV infections, including development of antiviral therapies and vaccines that target specific immune system antagonists within IECs.
Collapse
Affiliation(s)
- Joshua Oluoch Amimo
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Sergei Alekseevich Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Juliet Chepngeno
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Alfred Omwando Mainga
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Public Health, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Yusheng Guo
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Linda Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
15
|
Prates-Syed WA, Chaves LCS, Crema KP, Vuitika L, Lira A, Côrtes N, Kersten V, Guimarães FEG, Sadraeian M, Barroso da Silva FL, Cabral-Marques O, Barbuto JAM, Russo M, Câmara NOS, Cabral-Miranda G. VLP-Based COVID-19 Vaccines: An Adaptable Technology against the Threat of New Variants. Vaccines (Basel) 2021; 9:1409. [PMID: 34960155 PMCID: PMC8708688 DOI: 10.3390/vaccines9121409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/23/2022] Open
Abstract
Virus-like particles (VLPs) are a versatile, safe, and highly immunogenic vaccine platform. Recently, there are developmental vaccines targeting SARS-CoV-2, the causative agent of COVID-19. The COVID-19 pandemic affected humanity worldwide, bringing out incomputable human and financial losses. The race for better, more efficacious vaccines is happening almost simultaneously as the virus increasingly produces variants of concern (VOCs). The VOCs Alpha, Beta, Gamma, and Delta share common mutations mainly in the spike receptor-binding domain (RBD), demonstrating convergent evolution, associated with increased transmissibility and immune evasion. Thus, the identification and understanding of these mutations is crucial for the production of new, optimized vaccines. The use of a very flexible vaccine platform in COVID-19 vaccine development is an important feature that cannot be ignored. Incorporating the spike protein and its variations into VLP vaccines is a desirable strategy as the morphology and size of VLPs allows for better presentation of several different antigens. Furthermore, VLPs elicit robust humoral and cellular immune responses, which are safe, and have been studied not only against SARS-CoV-2 but against other coronaviruses as well. Here, we describe the recent advances and improvements in vaccine development using VLP technology.
Collapse
Affiliation(s)
- Wasim A. Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Lorena C. S. Chaves
- Department of Microbiology and Immunology, School of Medicine, Emory University, Claudia Nance Rollins Building, Atlanta, GA 30329, USA;
| | - Karin P. Crema
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Victor Kersten
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | | | - Mohammad Sadraeian
- São Carlos Institute of Physics, IFSC-USP, São Carlos 13566590, SP, Brazil; (F.E.G.G.); (M.S.)
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology, Sydney, NSW 2007, Australia
| | - Fernando L. Barroso da Silva
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040903, SP, Brazil;
- Department of Chemical and Biomolecular Engeneering, North Carolina State University, Raleigh, NC 27695, USA
| | - Otávio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508000, SP, Brazil
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children’s Medical Center, Tehran 1419733151, Iran
| | - José A. M. Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 0124690, SP, Brazil
| | - Momtchilo Russo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Niels O. S. Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| |
Collapse
|
16
|
Effect of GP19 Peptide Hyperimmune Antiserum on Activated Macrophage during Ehrlichia canis Infection in Canine Macrophage-like Cells. Animals (Basel) 2021; 11:ani11082310. [PMID: 34438767 PMCID: PMC8388390 DOI: 10.3390/ani11082310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022] Open
Abstract
In terms of its veterinary importance, vaccine development against Ehrlichia canis is needed. However, the effect of developing vaccines on humoral immune response against E. canis infection is still unknown. Novel GP194-43 was synthesized according to E. canis GP19 epitope prediction. To restrict any loss and/or illness in the host animal, rabbits were used in this study to produce GP194-43 hyperimmune sera. The effect of GP194-43 hyperimmune sera on neutralization was examined in vitro by determining the inhibition of E. canis infection of the macrophage-like cell line (DH82) in the presence of the sera. Four groups of DH82 cells received differing treatments. These included E. canis experimentally infected DH82 cells, E. canis-infected DH82 cells with control rabbit serum (untreated group), E. canis-infected DH82 cells with GP194-43 rabbit antiserum (treated group) and uninfected cells (negative control group), respectively. The treated group developed a decrease (p < 0.01) in the percentage of E. canis infected cells after 3 days post-infection at 48.57 ± 1.28. In addition, real-time PCR analyses of cytokine mRNA expression involved with the macrophage, humoral, and cellular immune responses were conducted. The findings revealed an upregulated expression of IFNG in the treated group during the infection. This study demonstrated neutralization in the GP194-43 peptide hyperimmune sera of immunized rabbits. Notably, IFN-γ production could be effectively promoted in canine macrophages in relation to the activation of macrophages and adaptive immune responses. The results of this study indicate the potential for the use of this immunogen in further investigations involving immunized and infected dogs as E. canis host species.
Collapse
|
17
|
Alves MLF, Ferreira MRA, Donassolo RA, Rodrigues RR, Conceição FR. Clostridium septicum: A review in the light of alpha-toxin and development of vaccines. Vaccine 2021; 39:4949-4956. [PMID: 34312008 DOI: 10.1016/j.vaccine.2021.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/31/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Clostridium septicum (CS) is a pathogen that can cause the death of animals in livestock worldwide through its main virulence factor, alpha-toxin (ATX). The aspects involved in diseases caused by ATX, such as economic impact, prevalence, and rapid clinical course, require that animals should be systematically immunized. This review provides an overview of CS in livestock farming and discusses current immunization methods. Currently, commercial vaccines available against CS involve the cultivation and inactivation of microorganisms and toxins using a time-consuming, expensive, and high biological risk-carrying production platform, and some have been reported to be ineffective. An alternative to this process is the recombinant DNA technology, although recombinant ATX obtained thus far is no longer efficient in stimulating protective antibody titers despite improvements in the production methods. On the other hand, immunized animals have highly favorable levels of survival when subjected to challenge tests, suggesting that high titers of circulating serum antibodies may not be representative of protection after immunization and that the non-immune cellular defenses associated with the particularities of the mechanism of action of ATX may be involved in the immune response of the host. To contribute to the future of global livestock farming through the development of more efficient recombinant vaccines, we suggest novel perspectives and strategies, such as the location of immunodominant epitopes, expression of relevant functional domains, and construction of chimeras, in the rational design of recombinant ATX.
Collapse
Affiliation(s)
- Mariliana Luiza Ferreira Alves
- Instituto Federal Sul-rio-grandense - IFSUL, Praça Vinte de Setembro, 455, Centro, CEP 96.015-360, Pelotas, RS, Brazil; Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, CEP 96160-000, Pelotas, RS, Brazil.
| | - Marcos Roberto Alves Ferreira
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, CEP 96160-000, Pelotas, RS, Brazil
| | - Rafael Amaral Donassolo
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, CEP 96160-000, Pelotas, RS, Brazil
| | - Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, CEP 96160-000, Pelotas, RS, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, CEP 96160-000, Pelotas, RS, Brazil
| |
Collapse
|
18
|
Ding P, Zhang G, Chen Y, Liu H, Liu Y, Jia R, Wang Y, Li G, Wang A. Reasonable permutation of M2e enhances the effect of universal influenza nanovaccine. Int J Biol Macromol 2021; 173:244-250. [PMID: 33485888 DOI: 10.1016/j.ijbiomac.2021.01.132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/25/2020] [Accepted: 01/19/2021] [Indexed: 01/19/2023]
Abstract
Influenza A virus (IAV) occasionally cross-species transmission among humans, swine and avian. The ectodomain of matrix protein 2 (M2e) is highly conserved in IAV, and multi-copy M2e from different species are usually displayed on the surface of nanoparticles to improve immunogenicity and constitute universal IAV nanovaccines. In our previous study, three M2e were inserted into the C-terminal of Cap protein of porcine circovirus type 2 (PCV2) to form a universal nanovaccine that provides protection against PCV2 and different subtypes of IAV. However, M2e adopts at least two converted conformations, and the intermolecular linker of M2e enhances the conformational instability, which limits the recognition by B cell receptors and production of high-level antibodies. Here, we report that the permutation of M2e affects effectiveness of nanovaccines. Three M2e derived from humans, swine and avian IAV were inserted into the C-terminal of Cap protein to form nanovaccines. Immunoprotective effects of different M2e arrangements were explored in mice. Results showed that the M2e closest to the surface of nanoparticle induced the most efficient protection against IAV derived from corresponding species. The results will contribute to develop more effective PCV2 and universal IAV bivalent nanovaccines for pigs, as well as species-specific universal IAV vaccines.
Collapse
Affiliation(s)
- Peiyang Ding
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Gaping Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongliang Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Yunchao Liu
- Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Rui Jia
- Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Yanwei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Ge Li
- Henan Zhongze Biological Engineering Co., Ltd., Zhengzhou 450002, China
| | - Aiping Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
19
|
Sarli M, Novoa MB, Mazzucco MN, Signorini ML, Echaide IE, de Echaide ST, Primo ME. A vaccine using Anaplasma marginale subdominant type IV secretion system recombinant proteins was not protective against a virulent challenge. PLoS One 2020; 15:e0229301. [PMID: 32084216 PMCID: PMC7034839 DOI: 10.1371/journal.pone.0229301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/03/2020] [Indexed: 12/26/2022] Open
Abstract
Anaplasma marginale is the most prevalent tick-borne livestock pathogen with worldwide distribution. Bovine anaplasmosis is a significant threat to cattle industry. Anaplasmosis outbreaks in endemic areas are prevented via vaccination with live A. centrale produced in splenectomized calves. Since A. centrale live vaccine can carry other pathogens and cause disease in adult cattle, research efforts are directed to develop safe recombinant subunit vaccines. Previous work found that the subdominant proteins of A. marginale type IV secretion system (T4SS) and the subdominant elongation factor-Tu (Ef-Tu) were involved in the protective immunity against the experimental challenge in cattle immunized with the A. marginale outer membrane (OM). This study evaluated the immunogenicity and protection conferred by recombinant VirB9.1, VirB9.2, VirB10, VirB11, and Ef-Tu proteins cloned and expressed in E. coli. Twenty steers were randomly clustered into four groups (G) of five animals each. Cattle from G1 and G2 were immunized with a mixture of 50 μg of each recombinant protein with Quil A® or Montanide™ adjuvants, respectively. Cattle from G3 and G4 (controls) were immunized with Quil A and Montanide adjuvants, respectively. Cattle received four immunizations at three-week intervals and were challenged with 107 A. marginale-parasitized erythrocytes 42 days after the fourth immunization. After challenge, all cattle showed clinical signs, with a significant drop of packed cell volume and a significant increase of parasitized erythrocytes (p<0.05), requiring treatment with oxytetracycline to prevent death. The levels of IgG2 induced in the immunized groups did not correlate with the observed lack of protection. Additional strategies are required to evaluate the role of these proteins and their potential utility in the development of effective vaccines.
Collapse
Affiliation(s)
- Macarena Sarli
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rafaela, Santa Fe, Argentina
| | - María B. Novoa
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rafaela, Santa Fe, Argentina
| | - Matilde N. Mazzucco
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
| | - Marcelo L. Signorini
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rafaela, Santa Fe, Argentina
| | - Ignacio E. Echaide
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
| | - Susana T. de Echaide
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
| | - María E. Primo
- Instituto Nacional de Tecnología Agropecuaria, Estación Experimental Agropecuaria Rafaela, Rafaela, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rafaela, Santa Fe, Argentina
| |
Collapse
|
20
|
Fernández-Prada M, López Trigo JA, Bayas JM, Cambronero MDR. [Quo vadis in vaccines: From the empirical approach to the new wave of technology]. Rev Esp Geriatr Gerontol 2020; 55:160-168. [PMID: 32014265 DOI: 10.1016/j.regg.2019.09.001] [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: 04/22/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 01/02/2023]
Abstract
The development of vaccines is a multifactorial process that has evolved and expanded, particularly over the last decades. The search for immunogenic vaccines that are also acceptably safe and tolerable enacted continuous technological advances in this field. In this regard, the technology applied to vaccines can historically be divided into 3 approaches: the empirical approach, the modern approach, and the new technological wave. The empirical approach for vaccine development includes whole micro-organisms, attenuation, inactivation, cell cultures and sub-unit vaccines. The modern approach contributed to leaps and bounds to vaccine development using chemical conjugation, as well as recombinant protein DNA technology and reverse vaccinology. Lastly, the new technological wave includes, among others, bioconjugation, viral vectors, synthetic biology, self-amplification of messenger RNA, generalized modules for membrane antigens, structural vaccinology and the new adjuvants.
Collapse
Affiliation(s)
- María Fernández-Prada
- Servicio de Medicina Preventiva y Salud Pública, Hospital Vital Álvarez Buylla, Mieres, España
| | | | - José M Bayas
- Departamento Médico, GSK, Tres Cantos, Madrid, España
| | | |
Collapse
|
21
|
Abstract
Enabled by new approaches for rapid identification and selection of human monoclonal antibodies, atomic-level structural information for viral surface proteins, and capacity for precision engineering of protein immunogens and self-assembling nanoparticles, a new era of antigen design and display options has evolved. While HIV-1 vaccine development has been a driving force behind these technologies and concepts, clinical proof-of-concept for structure-based vaccine design may first be achieved for respiratory syncytial virus (RSV), where conformation-dependent access to neutralization-sensitive epitopes on the fusion glycoprotein determines the capacity to induce potent neutralizing activity. Success with RSV has motivated structure-based stabilization of other class I viral fusion proteins for use as immunogens and demonstrated the importance of structural information for developing vaccines against other viral pathogens, particularly difficult targets that have resisted prior vaccine development efforts. Solving viral surface protein structures also supports rapid vaccine antigen design and application of platform manufacturing approaches for emerging pathogens.
Collapse
Affiliation(s)
- Barney S Graham
- Vaccine Research Center, National Institute of Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20850, USA;
| | - Morgan S A Gilman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA;
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, USA;
| |
Collapse
|
22
|
Oakes RS, Froimchuk E, Jewell CM. Engineering Biomaterials to Direct Innate Immunity. ADVANCED THERAPEUTICS 2019; 2:1800157. [PMID: 31236439 PMCID: PMC6590522 DOI: 10.1002/adtp.201800157] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Indexed: 12/18/2022]
Abstract
Small alterations during early stages of innate immune response can drive large changes in how adaptive immune cells develop and function during protective immunity or disease. Controlling these events creates exciting potential in development of immune engineered vaccines and therapeutics. This progress report discusses recent biomaterial technologies exploiting innate immunity to dissect immune function and to design new vaccines and immunotherapies for infectious diseases, cancer, and autoimmunity. Across these examples, an important idea is the possibility to co-opt innate immune mechanisms to enhance immunity during infection and cancer. During inflammatory or autoimmune disease, some of these same innate immune mechanisms can be manipulated in different ways to control excess inflammation by promotion of immunological tolerance.
Collapse
Affiliation(s)
- R. S. Oakes
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA
| | - E. Froimchuk
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA
| | - C. M. Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD, 20742, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, Maryland 21201, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
23
|
Majumder S, Das S, Somani VK, Makam SS, Kingston JJ, Bhatnagar R. A Bivalent Protein r-PAbxpB Comprising PA Domain IV and Exosporium Protein BxpB Confers Protection Against B. anthracis Spores and Toxin. Front Immunol 2019; 10:498. [PMID: 30941133 PMCID: PMC6433990 DOI: 10.3389/fimmu.2019.00498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 02/25/2019] [Indexed: 11/30/2022] Open
Abstract
Anthrax vaccines primarily relying only on protective antigen (PA), the cell binding component in anthrax toxins provide incomplete protection when challenged with spores of virulent encapsulated Bacillus anthracis strains. Alternatively, formaldehyde inactivated spores (FIS) or recombinant spore components generate anti-spore immune responses that inhibit the early stages of infection and augment the PA protective efficacy. In the present study domain IV of PA was spliced with exosporium antigen BxpB via a flexible G4S linker to generate a single functional antigen r-PAbxpB that was further assessed for its protective efficacy against anthrax toxins and spore infection. Immunization of mice with r-PAbxpB elicited significantly high titer antibodies comprising IgG1:IgG2a isotypes in 1:1 ratio, balanced up-regulation of both Th1 (IL2, IL12, IFN-γ) and Th2 (IL4, IL5, IL10) cytokines and high frequencies of CD4+ and CD8+ T cell subsets. The anti-r-PAbxpB antibodies significantly enhanced spore phagocytosis, and killing within macrophages; inhibited their germination to vegetative cells and completely neutralized the anthrax toxins as evidenced by the 100% protection in passive transfer studies. Active immunization with r-PAbxpB provided 100 and 83.3% protection in mice I.P. challenged with 5 × LD100 LD of toxins and 5 × 104 cfu/ml Ames spores, respectively while the sham immunized group succumbed to infection in 48 h. Therefore, the ability of r-PAbxpB to generate protective immune responses against both spores and toxin and provide significant protection suggests it as an efficient vaccine candidate against B. anthracis infection.
Collapse
Affiliation(s)
- Saugata Majumder
- Defence Food Research Laboratory, Microbiology Division, Defence Research Development Organisation, Mysore, India
| | - Shreya Das
- Defence Food Research Laboratory, Microbiology Division, Defence Research Development Organisation, Mysore, India
| | | | - Shivakiran S Makam
- Defence Food Research Laboratory, Microbiology Division, Defence Research Development Organisation, Mysore, India
| | - Joseph J Kingston
- Defence Food Research Laboratory, Microbiology Division, Defence Research Development Organisation, Mysore, India
| | - Rakesh Bhatnagar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
24
|
Abstract
Although vaccines have been successfully developed against several pathogens, designing an effective vaccine to protect against human immunodeficiency virus (HIV) has remained an intractable challenge. To address this, the research community has looked to human and non-human primate studies to understand the correlates of protective immunity, based on which a targeted vaccine strategy may be designed. Two distinct approaches, focused on different immune correlates of protection, have emerged. The first focuses on structure-based design of HIV envelope immunogens that are able to induce antibodies that neutralize the virus. The second focuses on strategies aimed at driving non-neutralizing polyclonal and polyfunctional antibodies that engage other arms of immunity to clear the virus. Here we review these two different vaccine design strategies and posit that ultimately the convergence of these two efforts will likely be necessary for the development of a globally protective HIV vaccine.
Collapse
Affiliation(s)
- Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA.
| | - Dan Barouch
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Bidmos FA, Siris S, Gladstone CA, Langford PR. Bacterial Vaccine Antigen Discovery in the Reverse Vaccinology 2.0 Era: Progress and Challenges. Front Immunol 2018; 9:2315. [PMID: 30349542 PMCID: PMC6187972 DOI: 10.3389/fimmu.2018.02315] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/17/2018] [Indexed: 11/13/2022] Open
Abstract
The ongoing, and very serious, threat from antimicrobial resistance necessitates the development and use of preventative measures, predominantly vaccination. Polysaccharide-based vaccines have provided a degree of success in limiting morbidity from disseminated bacterial infections, including those caused by the major human obligate pathogens, Neisseria meningitidis, and Streptococcus pneumoniae. Limitations of these polysaccharide vaccines, such as partial coverage and induced escape leading to persistence of disease, provide a compelling argument for the development of protein vaccines. In this review, we briefly chronicle approaches that have yielded licensed vaccines before highlighting reverse vaccinology 2.0 and its potential application in the discovery of novel bacterial protein vaccine candidates. Technical challenges and research gaps are also discussed.
Collapse
Affiliation(s)
- Fadil A Bidmos
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Sara Siris
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Paul R Langford
- Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Ferraro M, Colombo G. Targeting Difficult Protein-Protein Interactions with Plain and General Computational Approaches. Molecules 2018; 23:molecules23092256. [PMID: 30181519 PMCID: PMC6225287 DOI: 10.3390/molecules23092256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 11/21/2022] Open
Abstract
Investigating protein-protein interactions (PPIs) holds great potential for therapeutic applications, since they mediate intricate cell signaling networks in physiological and disease states. However, their complex and multifaceted nature poses a major challenge for biochemistry and medicinal chemistry, thereby limiting the druggability of biological partners participating in PPIs. Molecular Dynamics (MD) provides a solid framework to study the reciprocal shaping of proteins’ interacting surfaces. Here, we review successful applications of MD-based methods developed in our group to predict interfacial areas involved in PPIs of pharmaceutical interest. We report two interesting examples of how structural, dynamic and energetic information can be combined into efficient strategies which, complemented by experiments, can lead to the design of new small molecules with promising activities against cancer and infections. Our advances in targeting key PPIs in angiogenic pathways and antigen-antibody recognition events will be discussed for their role in drug discovery and chemical biology.
Collapse
Affiliation(s)
- Mariarosaria Ferraro
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Via Mario Bianco 9, 20131 Milano, Italy.
| | - Giorgio Colombo
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Via Mario Bianco 9, 20131 Milano, Italy.
- Dipartimento di Chimica, Università di Pavia, V.le Taramelli 10, 27100 Pavia, Italy.
| |
Collapse
|
27
|
Bavaro T, Pinto A, Dall’Oglio F, Hernáiz MJ, Morelli CF, Zambelli P, De Micheli C, Conti P, Tamborini L, Terreni M. Flow-based biocatalysis: Application to peracetylated arabinofuranosyl-1,5-arabinofuranose synthesis. Process Biochem 2018. [DOI: 10.1016/j.procbio.2018.06.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Postgenomic Approaches and Bioinformatics Tools to Advance the Development of Vaccines against Bacteria of the Burkholderia cepacia Complex. Vaccines (Basel) 2018; 6:vaccines6020034. [PMID: 29890657 PMCID: PMC6027386 DOI: 10.3390/vaccines6020034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022] Open
Abstract
Bacteria of the Burkholderia cepacia complex (Bcc) remain an important cause of morbidity and mortality among patients suffering from cystic fibrosis. Eradication of these pathogens by antimicrobial therapy often fails, highlighting the need to develop novel strategies to eradicate infections. Vaccines are attractive since they can confer protection to particularly vulnerable patients, as is the case of cystic fibrosis patients. Several studies have identified specific virulence factors and proteins as potential subunit vaccine candidates. So far, no vaccine is available to protect from Bcc infections. In the present work, we review the most promising postgenomic approaches and selected web tools available to speed up the identification of immunogenic proteins with the potential of conferring protection against Bcc infections.
Collapse
|
29
|
Codon optimization and improved delivery/immunization regimen enhance the immune response against wild-type and drug-resistant HIV-1 reverse transcriptase, preserving its Th2-polarity. Sci Rep 2018; 8:8078. [PMID: 29799015 PMCID: PMC5967322 DOI: 10.1038/s41598-018-26281-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
DNA vaccines require a considerable enhancement of immunogenicity. Here, we optimized a prototype DNA vaccine against drug-resistant HIV-1 based on a weak Th2-immunogen, HIV-1 reverse transcriptase (RT). We designed expression-optimized genes encoding inactivated wild-type and drug-resistant RTs (RT-DNAs) and introduced them into mice by intradermal injections followed by electroporation. RT-DNAs were administered as single or double primes with or without cyclic-di-GMP, or as a prime followed by boost with RT-DNA mixed with a luciferase-encoding plasmid (“surrogate challenge”). Repeated primes improved cellular responses and broadened epitope specificity. Addition of cyclic-di-GMP induced a transient increase in IFN-γ production. The strongest anti-RT immune response was achieved in a prime-boost protocol with electroporation by short 100V pulses done using penetrating electrodes. The RT-specific response, dominated by CD4+ T-cells, targeted epitopes at aa 199–220 and aa 528–543. Drug-resistance mutations disrupted the epitope at aa 205–220, while the CTL epitope at aa 202–210 was not affected. Overall, multiparametric optimization of RT strengthened its Th2- performance. A rapid loss of RT/luciferase-expressing cells in the surrogate challenge experiment revealed a lytic potential of anti-RT response. Such lytic CD4+ response would be beneficial for an HIV vaccine due to its comparative insensitivity to immune escape.
Collapse
|
30
|
Minz S, Pandey RS. Lipid A adjuvanted Chylomicron Mimicking Solid Fat Nanoemulsions for Immunization Against Hepatitis B. AAPS PharmSciTech 2018; 19:1168-1181. [PMID: 29243216 DOI: 10.1208/s12249-017-0932-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/27/2017] [Indexed: 11/30/2022] Open
Abstract
Traditional parenteral recombinant hepatitis B virus (HBV) vaccines have effectively reduced the disease burden despite being able to induce seroprotective antibody titers in 5-10% vaccinated individuals (non-responders). Moreover, an estimated 340 million chronic HBV cases are in need of treatment. Development of safe, stable, and more effective hepatitis B vaccine formulation would address these challenges. Recombinant hepatitis B surface antigen (rHBsAg) entrapped solid fat nanoemulsions (SFNs) containing monophosphoryl lipid A (MPLA) that was prepared and optimized by quality by design (QbD) using response surface methodology (RSM), i.e., central composite design (CCD). Its immune potential was evaluated with preset immunization protocol in a murine model. Dose escalation study revealed that formulation containing 1 μg of rHBsAg entrapped SFNs with MPLA-induced significant higher humoral, and cellular response compared to the marketed vaccine (Genvac B) administered intramuscularly. SFNs with nanometric morphology and structural similarity with chylomicrons assist in improved uptake and processing to lymphatics. Moreover, the presence of an immunogenic component in its structure further augments delivery of rHBsAg to immune cells with induction of danger signals. This multi-adjuvant based approach explores new prospect for the dose sparing. Improved cellular immune response induced by this vaccine formulation suggests that it could be tested as an immunotherapeutic vaccine as well.
Collapse
|
31
|
Cyrklaff M, Frischknecht F, Kudryashev M. Functional insights into pathogen biology from 3D electron microscopy. FEMS Microbiol Rev 2018; 41:828-853. [PMID: 28962014 DOI: 10.1093/femsre/fux041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/25/2017] [Indexed: 01/10/2023] Open
Abstract
In recent years, novel imaging approaches revolutionised our understanding of the cellular and molecular biology of microorganisms. These include advances in fluorescent probes, dynamic live cell imaging, superresolution light and electron microscopy. Currently, a major transition in the experimental approach shifts electron microscopy studies from a complementary technique to a method of choice for structural and functional analysis. Here we review functional insights into the molecular architecture of viruses, bacteria and parasites as well as interactions with their respective host cells gained from studies using cryogenic electron tomography and related methodologies.
Collapse
Affiliation(s)
- Marek Cyrklaff
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Mikhail Kudryashev
- Max Planck Institute of Biophysics, Max-von-Laue Strasse 3, 60438 Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University of Frankfurt, Max-von-Laue Strasse 17, 60438 Frankfurt, Germany
| |
Collapse
|
32
|
Hsieh CL, Ptak CP, Tseng A, Suguiura IMDS, McDonough SP, Sritrakul T, Li T, Lin YP, Gillilan RE, Oswald RE, Chang YF. Extended low-resolution structure of a Leptospira antigen offers high bactericidal antibody accessibility amenable to vaccine design. eLife 2017; 6:e30051. [PMID: 29210669 PMCID: PMC5749957 DOI: 10.7554/elife.30051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/02/2017] [Indexed: 01/16/2023] Open
Abstract
Pathogens rely on proteins embedded on their surface to perform tasks essential for host infection. These obligatory structures exposed to the host immune system provide important targets for rational vaccine design. Here, we use a systematically designed series of multi-domain constructs in combination with small angle X-ray scattering (SAXS) to determine the structure of the main immunoreactive region from a major antigen from Leptospira interrogans, LigB. An anti-LigB monoclonal antibody library exhibits cell binding and bactericidal activity with extensive domain coverage complementing the elongated architecture observed in the SAXS structure. Combining antigenic motifs in a single-domain chimeric immunoglobulin-like fold generated a vaccine that greatly enhances leptospiral protection over vaccination with single parent domains. Our study demonstrates how understanding an antigen's structure and antibody accessible surfaces can guide the design and engineering of improved recombinant antigen-based vaccines.
Collapse
Affiliation(s)
- Ching-Lin Hsieh
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | - Christopher P Ptak
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
- Department of Molecular Medicine, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | - Andrew Tseng
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | | | - Sean P McDonough
- Department of Biomedical Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | - Tepyuda Sritrakul
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | - Ting Li
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | - Yi-Pin Lin
- Division of Infectious DiseaseWadsworth Center, New York State Department of HealthAlbanyUnited States
| | - Richard E Gillilan
- Macromolecular Diffraction Facility at CHESS (MacCHESS)Cornell UniversityIthacaUnited States
| | - Robert E Oswald
- Department of Molecular Medicine, College of Veterinary MedicineCornell UniversityIthacaUnited States
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary MedicineCornell UniversityIthacaUnited States
| |
Collapse
|
33
|
Brady JM, Baltimore D, Balazs AB. Antibody gene transfer with adeno-associated viral vectors as a method for HIV prevention. Immunol Rev 2017; 275:324-333. [PMID: 28133808 DOI: 10.1111/imr.12478] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Broadly neutralizing antibodies (bNAbs) against human immunodeficiency virus (HIV) show great promise in HIV prevention as they are capable of potently neutralizing a considerable breadth of genetically diverse strains. Passive transfer of monoclonal bNAb proteins can confer protection in animal models of HIV infection at modest concentrations, inspiring efforts to develop an HIV vaccine capable of eliciting bNAb responses. However, these antibodies demonstrate high degrees of somatic mutation and other unique characteristics that may hinder the ability of conventional approaches to consistently and effectively produce bNAb analogs. As an alternative strategy, we and others have proposed vector-mediated gene transfer to generate long-term, systemic production of bNAbs in the absence of immunization. Herein, we review the use of adeno-associated virus (AAV) vectors for delivery of HIV bNAbs and antibody-like proteins and summarize both the advantages and disadvantages of this strategy as a method for HIV prevention.
Collapse
Affiliation(s)
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | |
Collapse
|
34
|
Wang L, Xing D, Le Van A, Jerse AE, Wang S. Structure-based design of ferritin nanoparticle immunogens displaying antigenic loops of Neisseria gonorrhoeae. FEBS Open Bio 2017; 7:1196-1207. [PMID: 28781959 PMCID: PMC5537070 DOI: 10.1002/2211-5463.12267] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 11/09/2022] Open
Abstract
Effective vaccines are urgently needed to combat gonorrhea, a common sexually transmitted bacterial infection, for which treatment options are diminishing due to rapid emergence of antibiotic resistance. We have used a rational approach to the development of gonorrhea vaccines, and genetically engineered nanoparticles to present antigenic peptides of Neisseria gonorrhoeae, the causative agent of gonorrhea. We hypothesized that the ferritin nanocage could be used as a platform to display an ordered array of N. gonorrhoeae antigenic peptides on its surface. MtrE, the outer membrane channel of the highly conserved gonococcal MtrCDE active efflux pump, is an attractive vaccine target due to its importance in protecting N. gonorrhoeae from host innate effectors and antibiotic resistance. Using computational approaches, we designed constructs that expressed chimeric proteins of the Helicobacter pylori ferritin and antigenic peptides that correspond to the two surface-exposed loops of N. gonorrhoeae MtrE. The peptides were inserted at the N terminus or in a surface-exposed ferritin loop between helices αA and αB. Crystal structures of the chimeric proteins revealed that the proteins assembled correctly into a 24-mer nanocage structure. Although the inserted N. gonorrhoeae peptides were disordered, it was clear that they were displayed on the nanocage surface, but with multiple conformations. Our results confirmed that the ferritin nanoparticle is a robust platform to present antigenic peptides and therefore an ideal system for rational design of immunogens.
Collapse
Affiliation(s)
- Liqin Wang
- Department of Biochemistry and Molecular Biology Uniformed Services University of the Health Sciences Bethesda MD USA
| | - Daniel Xing
- Department of Biochemistry and Molecular Biology Uniformed Services University of the Health Sciences Bethesda MD USA
| | - Adriana Le Van
- Department of Microbiology and Immunology Uniformed Services University of the Health Sciences Bethesda MD USA
| | - Ann E Jerse
- Department of Microbiology and Immunology Uniformed Services University of the Health Sciences Bethesda MD USA
| | - Shuishu Wang
- Department of Biochemistry and Molecular Biology Uniformed Services University of the Health Sciences Bethesda MD USA
| |
Collapse
|
35
|
Ding P, Zhang T, Li Y, Teng M, Sun Y, Liu X, Chai S, Zhou E, Jin Q, Zhang G. Nanoparticle orientationally displayed antigen epitopes improve neutralizing antibody level in a model of porcine circovirus type 2. Int J Nanomedicine 2017; 12:5239-5254. [PMID: 28769561 PMCID: PMC5533572 DOI: 10.2147/ijn.s140789] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent advancements in biotechnology have enabled the rapid identification and subsequent expression of pathogenic microbial major antigens that induce protective immune responses. However, subunit vaccines have not been successfully commercialized mainly due to the lack of sufficient levels of neutralizing antibodies (NAs). High levels of NA rely on the efficient recognition and cross-linking of multiple neutralizing epitopes with B-cell receptors (BCRs). Nanoparticles are able to display coupled antigenic arrays at high density and provide multiple binding molecular scenarios with BCRs. The high-resolution antigenic structure makes it possible to accurately display stable neutralizing epitopes. Therefore, the development of a nanovaccine that orientationally displays neutralizing epitopes is a feasible strategy. To address this hypothesis, the capsid (Cap) protein of porcine circovirus type 2 as model antigen was conjugated to gold nanoparticles (AuNPs) through direct reaction of the mercapto group of the unique cysteines with AuNPs, rendering Cap-AuNPs to have neutralizing epitopes on outer surface and an immunodominant epitope buried within the inner surface. In vitro studies showed that AuNPs promoted the phagocytosis of Cap protein and NA levels were significantly improved, meanwhile antibody levels against the immunodominant epitope was significantly reduced. In mouse studies, Cap-AuNP-immunized mice displayed a high production of interleukin (IL)-4, IL-10, and interferon-γ, suggesting that Cap-AuNPs can effectively activate CD4+ and CD8+ T cells and balance Th1 and Th2 cellular responses. This study presents a new vaccine design strategy based on antigen structure, where nanoparticles are coupled to antigens in well-ordered arrays and orientationally display neutralizing epitopes to enhance NA levels.
Collapse
Affiliation(s)
- Peiyang Ding
- College of Veterinary Medicine, Northwest A&F University, Yangling.,Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Teng Zhang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,College of Life Sciences, Henan Agricultural University
| | - Yafei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling.,Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Man Teng
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Yaning Sun
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Xiao Liu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou
| | - Shujun Chai
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Enmin Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling
| | - Qianyue Jin
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling.,Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
36
|
Paladino A, Marchetti F, Rinaldi S, Colombo G. Protein design: from computer models to artificial intelligence. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2017. [DOI: 10.1002/wcms.1318] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Antonella Paladino
- Biomolecular Simulations & Computational Chemistry Group; Istituto Istituto di Chimica del Riconoscimento Molecolare, CNR; Milano Italy
| | - Filippo Marchetti
- Biomolecular Simulations & Computational Chemistry Group; Istituto Istituto di Chimica del Riconoscimento Molecolare, CNR; Milano Italy
| | - Silvia Rinaldi
- Biomolecular Simulations & Computational Chemistry Group; Istituto Istituto di Chimica del Riconoscimento Molecolare, CNR; Milano Italy
| | - Giorgio Colombo
- Biomolecular Simulations & Computational Chemistry Group; Istituto Istituto di Chimica del Riconoscimento Molecolare, CNR; Milano Italy
| |
Collapse
|
37
|
Favuzza P, Guffart E, Tamborrini M, Scherer B, Dreyer AM, Rufer AC, Erny J, Hoernschemeyer J, Thoma R, Schmid G, Gsell B, Lamelas A, Benz J, Joseph C, Matile H, Pluschke G, Rudolph MG. Structure of the malaria vaccine candidate antigen CyRPA and its complex with a parasite invasion inhibitory antibody. eLife 2017; 6. [PMID: 28195038 PMCID: PMC5349852 DOI: 10.7554/elife.20383] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/06/2017] [Indexed: 12/02/2022] Open
Abstract
Invasion of erythrocytes by Plasmodial merozoites is a composite process involving the interplay of several proteins. Among them, the Plasmodium falciparum Cysteine-Rich Protective Antigen (PfCyRPA) is a crucial component of a ternary complex, including Reticulocyte binding-like Homologous protein 5 (PfRH5) and the RH5-interacting protein (PfRipr), essential for erythrocyte invasion. Here, we present the crystal structures of PfCyRPA and its complex with the antigen-binding fragment of a parasite growth inhibitory antibody. PfCyRPA adopts a 6-bladed β-propeller structure with similarity to the classic sialidase fold, but it has no sialidase activity and fulfills a purely non-enzymatic function. Characterization of the epitope recognized by protective antibodies may facilitate design of peptidomimetics to focus vaccine responses on protective epitopes. Both in vitro and in vivo anti-PfCyRPA and anti-PfRH5 antibodies showed more potent parasite growth inhibitory activity in combination than on their own, supporting a combined delivery of PfCyRPA and PfRH5 in vaccines. DOI:http://dx.doi.org/10.7554/eLife.20383.001 Malaria is one of the deadliest infectious diseases worldwide, killing over 400,000 people a year. About 200 million people are infected every year, placing a huge social and medical burden especially on developing countries. Microscopic parasites known as Plasmodium are responsible for causing this disease. Plasmodium parasites have a complex life cycle involving both mosquito and mammal hosts. This includes a stage where the parasites infect the mammal’s red blood cells, which causes the symptoms of the disease. In 2012, a team of researchers discovered that a protein called CyRPA forms a group (or ‘complex’) with several other proteins to allow the parasites to enter red blood cells. Developing a vaccine is one of the most promising approaches to prevent malaria. Vaccines help the body to recognise and fight an invading microbe by triggering an immune response that results in the production of proteins called antibodies, which can bind to specific molecules on the surface of the microbe. If the microbe later enters the body, these antibodies can be produced quickly to eliminate the microbe before it causes disease. However, efforts to develop a highly effective vaccine against malaria have so far been unsuccessful. Favuzza et al. – including some of the researchers involved in the 2012 work – used a technique called X-ray crystallography to investigate the three-dimensional structure of the CyRPA protein. The experiments show that an antibody is able to bind to a region of CyRPA – a designated ‘protective epitope’ – that is similar in the CyRPA proteins of all Plasmodium falciparum strains. These antibodies can prevent the parasite from entering the red blood cells, and vaccines containing CyRPA may therefore be effective at protecting individuals from malaria. The findings of Favuzza et al. also suggest that using CyRPA in combination with another protein in the complex called RH5 could make the vaccine more powerful as it would make it harder for the parasite to become resistant. The next step following on from this work is to design a vaccine containing protective CyRPA epitopes that triggers an immune response in mammals that is strong enough to reduce the numbers of parasites in the blood. A future challenge will be to develop a vaccine that combines several proteins involved in different stages of the parasite’s life cycle to provide full protection against malaria. DOI:http://dx.doi.org/10.7554/eLife.20383.002
Collapse
Affiliation(s)
- Paola Favuzza
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Elena Guffart
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Marco Tamborrini
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Bianca Scherer
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Anita M Dreyer
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Arne C Rufer
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Johannes Erny
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Joerg Hoernschemeyer
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ralf Thoma
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Georg Schmid
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Bernard Gsell
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Araceli Lamelas
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Joerg Benz
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Catherine Joseph
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Hugues Matile
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Gerd Pluschke
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Markus G Rudolph
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
38
|
Du L, Tai W, Yang Y, Zhao G, Zhu Q, Sun S, Liu C, Tao X, Tseng CTK, Perlman S, Jiang S, Zhou Y, Li F. Introduction of neutralizing immunogenicity index to the rational design of MERS coronavirus subunit vaccines. Nat Commun 2016; 7:13473. [PMID: 27874853 PMCID: PMC5121417 DOI: 10.1038/ncomms13473] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/06/2016] [Indexed: 12/13/2022] Open
Abstract
Viral subunit vaccines often contain immunodominant non-neutralizing epitopes that divert host immune responses. These epitopes should be eliminated in vaccine design, but there is no reliable method for evaluating an epitope's capacity to elicit neutralizing immune responses. Here we introduce a new concept 'neutralizing immunogenicity index' (NII) to evaluate an epitope's neutralizing immunogenicity. To determine the NII, we mask the epitope with a glycan probe and then assess the epitope's contribution to the vaccine's overall neutralizing immunogenicity. As proof-of-concept, we measure the NII for different epitopes on an immunogen comprised of the receptor-binding domain from MERS coronavirus (MERS-CoV). Further, we design a variant form of this vaccine by masking an epitope that has a negative NII score. This engineered vaccine demonstrates significantly enhanced efficacy in protecting transgenic mice from lethal MERS-CoV challenge. Our study may guide the rational design of highly effective subunit vaccines to combat MERS-CoV and other life-threatening viruses.
Collapse
Affiliation(s)
- Lanying Du
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York 10065, USA
| | - Wanbo Tai
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York 10065, USA
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yang Yang
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qing Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Shihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Chang Liu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | - Xinrong Tao
- Department of Microbiology and Immunology and Center for Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Chien-Te K. Tseng
- Department of Microbiology and Immunology and Center for Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Stanley Perlman
- Department of Microbiology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Shibo Jiang
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York 10065, USA
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
39
|
Hassan A, Naz A, Obaid A, Paracha RZ, Naz K, Awan FM, Muhmmad SA, Janjua HA, Ahmad J, Ali A. Pangenome and immuno-proteomics analysis of Acinetobacter baumannii strains revealed the core peptide vaccine targets. BMC Genomics 2016; 17:732. [PMID: 27634541 PMCID: PMC5025611 DOI: 10.1186/s12864-016-2951-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/19/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Acinetobacter baumannii has emerged as a significant nosocomial pathogen during the last few years, exhibiting resistance to almost all major classes of antibiotics. Alternative treatment options such as vaccines tend to be most promising and cost effective approaches against this resistant pathogen. In the current study, we have explored the pan-genome of A. baumannii followed by immune-proteomics and reverse vaccinology approaches to identify potential core vaccine targets. RESULTS The pan-genome of all available A. baumannii strains (30 complete genomes) is estimated to contain 7,606 gene families and the core genome consists of 2,445 gene families (~32 % of the pan-genome). Phylogenetic tree, comparative genomic and proteomic analysis revealed both intra- and inter genomic similarities and evolutionary relationships. Among the conserved core genome, thirteen proteins, including P pilus assembly protein, pili assembly chaperone, AdeK, PonA, OmpA, general secretion pathway protein D, FhuE receptor, Type VI secretion system OmpA/MotB, TonB dependent siderophore receptor, general secretion pathway protein D, outer membrane protein, peptidoglycan associated lipoprotein and peptidyl-prolyl cis-trans isomerase are identified as highly antigenic. Epitope mapping of the target proteins revealed the presence of antigenic surface exposed 9-mer T-cell epitopes. Protein-protein interaction and functional annotation have shown their involvement in significant biological and molecular processes. The pipeline is validated by predicting already known immunogenic targets against Gram negative pathogen Helicobacter pylori as a positive control. CONCLUSION The study, based upon combinatorial approach of pan-genomics, core genomics, proteomics and reverse vaccinology led us to find out potential vaccine candidates against A. baumannii. The comprehensive analysis of all the completely sequenced genomes revealed thirteen putative antigens which could elicit substantial immune response. The integration of computational vaccinology strategies would facilitate in tackling the rapid dissemination of resistant A.baumannii strains. The scarcity of effective antibiotics and the global expansion of sequencing data making this approach desirable in the development of effective vaccines against A. baumannii and other bacterial pathogens.
Collapse
Affiliation(s)
- Afreenish Hassan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Anam Naz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Ayesha Obaid
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Rehan Zafar Paracha
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Kanwal Naz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Faryal Mehwish Awan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Syed Aun Muhmmad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Hussnain Ahmed Janjua
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Jamil Ahmad
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
- Department of Computer Science and Information Technology, Stratford University, Falls Church, VA 22043 USA
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| |
Collapse
|
40
|
Wen X, Pickens J, Mousa JJ, Leser GP, Lamb RA, Crowe JE, Jardetzky TS. A Chimeric Pneumovirus Fusion Protein Carrying Neutralizing Epitopes of Both MPV and RSV. PLoS One 2016; 11:e0155917. [PMID: 27224013 PMCID: PMC4880302 DOI: 10.1371/journal.pone.0155917] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/06/2016] [Indexed: 11/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (HMPV) are paramyxoviruses that are responsible for substantial human health burden, particularly in children and the elderly. The fusion (F) glycoproteins are major targets of the neutralizing antibody response and studies have mapped dominant antigenic sites in F. Here we grafted a major neutralizing site of RSV F, recognized by the prophylactic monoclonal antibody palivizumab, onto HMPV F, generating a chimeric protein displaying epitopes of both viruses. We demonstrate that the resulting chimeric protein (RPM-1) is recognized by both anti-RSV and anti-HMPV F neutralizing antibodies indicating that it can be used to map the epitope specificity of antibodies raised against both viruses. Mice immunized with the RPM-1 chimeric antigen generate robust neutralizing antibody responses to MPV but weak or no cross-reactive recognition of RSV F, suggesting that grafting of the single palivizumab epitope stimulates a comparatively limited antibody response. The RPM-1 protein provides a new tool for characterizing the immune responses resulting from RSV and HMPV infections and provides insights into the requirements for developing a chimeric subunit vaccine that could induce robust and balanced immunity to both virus infections.
Collapse
Affiliation(s)
- Xiaolin Wen
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Jennifer Pickens
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Monroe Carell Jr. Children's Hospital at Vanderbilt University, Nashville, TN, United States of America
| | - Jarrod J. Mousa
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Monroe Carell Jr. Children's Hospital at Vanderbilt University, Nashville, TN, United States of America
| | - George P. Leser
- Howard Hughes Medical Institute, Northwestern University, Evanston, IL, United States of America
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
| | - Robert A. Lamb
- Howard Hughes Medical Institute, Northwestern University, Evanston, IL, United States of America
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
| | - James E. Crowe
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Monroe Carell Jr. Children's Hospital at Vanderbilt University, Nashville, TN, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Theodore S. Jardetzky
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
41
|
Kundu P, Biswas R, Mukherjee S, Reinhard L, Dutta A, Mueller-Dieckmann J, Weiss MS, Pal NK, Das AK. Structure-based Epitope Mapping of Mycobacterium tuberculosis Secretary Antigen MTC28. J Biol Chem 2016; 291:13943-13954. [PMID: 27189947 DOI: 10.1074/jbc.m116.726422] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Indexed: 11/06/2022] Open
Abstract
Secretary proteins of Mycobacterium tuberculosis are key players of the mycobacterial infection pathway. MTC28 is a 28-kDa proline-rich secretary antigen of Mycobacterium tuberculosis and is only conserved in pathogenic strains of mycobacteria. Here we report the crystal structure of MTC28 at 2.8- and 2.15-Å resolutions for the structure-based epitope design. MTC28 shares a "mog1p"-fold consisting of seven antiparallel β strands stacked between α helices. Five probable epitopes have been located on a solvent-accessible flexible region by computational analysis of the structure of MTC28. Simultaneously, the protein is digested with trypsin and the resulting fragments are purified by HPLC. Such 10 purified peptide fragments are screened against sera from patients infected with pulmonary tuberculosis (PTB). Two of these 10 fragments, namely (127)ALDITLPMPPR(137) and (138)WTQVPDPNVPDAFVVIADR(156),are found to be major immunogenic epitopes that are localized on the outer surface of the protein molecule and are part of a single continuous epitope that have been predicted in silico Mutagenesis and antibody inhibition studies are in accordance with the results obtained from epitope mapping.
Collapse
Affiliation(s)
- Prasun Kundu
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Rupam Biswas
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Somnath Mukherjee
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Linda Reinhard
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden,; Röntgen-Ångström-Cluster, Karolinska Institutet Outstation, Centre for Structural Systems Biology, c/o DESY, D-22607 Hamburg, Germany,; European Molecular Biology Laboratory, Hamburg Unit, 22603 Hamburg, Germany
| | - Anirudha Dutta
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Jochen Mueller-Dieckmann
- Abteilung für Mikrobiologie und Biotechnologie, Biozentrum Klein Flottbek, Universität Hamburg, Ohnhorststraße18, 22609 Hamburg, Germany
| | - Manfred S Weiss
- Macromolecular Crystallography (HZB-MX), Helmholtz-Zentrum Berlin für Materialien und Energie, D-12489 Berlin, Germany
| | - Nishit Kumar Pal
- Nil Ratan Sirkar Medical College and Hospital,138 AJC Bose Road, Sealdah, Raja Bazar, Kolkata, West Bengal 700014, India
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India,.
| |
Collapse
|
42
|
Abstract
Traditional vaccination with whole pathogens or pathogen-derived subunits has completely eliminated diseases like smallpox, and has greatly limited the incidence, morbidity and mortality associated with many other infectious diseases. Unfortunately, a large burden of infectious disease remains that may be preventable through vaccination. For many of these, more focused and innovative approaches may be essential for the development of effective vaccines.
Collapse
Affiliation(s)
- Jon Oscherwitz
- a Division of Hematology-Oncology, Department of Internal Medicine , University of Michigan Medical School , Ann Arbor , MI , USA.,b Veterans Administration Ann Arbor Healthcare System , Ann Arbor , MI , USA
| |
Collapse
|
43
|
Rappuoli R, Bottomley MJ, D'Oro U, Finco O, De Gregorio E. Reverse vaccinology 2.0: Human immunology instructs vaccine antigen design. J Exp Med 2016; 213:469-81. [PMID: 27022144 PMCID: PMC4821650 DOI: 10.1084/jem.20151960] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
Traditionally, vaccines have been developed by cultivating infectious agents and isolating the inactivated whole pathogen or some of its purified components. 20 years ago, reverse vaccinology enabled vaccine discovery and design based on information deriving from the sequence of microbial genomes rather than via the growth of pathogens. Today, the high throughput discovery of protective human antibodies, sequencing of the B cell repertoire, and the increasing structural characterization of protective antigens and epitopes provide the molecular and mechanistic understanding to drive the discovery of novel vaccines that were previously impossible. We are entering a "reverse vaccinology 2.0" era.
Collapse
Affiliation(s)
| | | | - Ugo D'Oro
- GlaxoSmithKline Vaccines S.r.l., 53100 Siena, Italy
| | - Oretta Finco
- GlaxoSmithKline Vaccines S.r.l., 53100 Siena, Italy
| | | |
Collapse
|
44
|
Geissner A, Pereira CL, Leddermann M, Anish C, Seeberger PH. Deciphering Antigenic Determinants of Streptococcus pneumoniae Serotype 4 Capsular Polysaccharide using Synthetic Oligosaccharides. ACS Chem Biol 2016; 11:335-44. [PMID: 26674834 DOI: 10.1021/acschembio.5b00768] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Streptococcus pneumoniae is a major cause of mortality and morbidity worldwide. More than 90 S. pneumoniae serotypes are distinguished based on the structure of their primary targets to the human immune system, the capsular polysaccharides (CPSs). The CPS of the prevalent serotype 4 (ST4) is composed of tetrasaccharide repeating units and is included in existing pneumococcal vaccines. Still, the structural antigenic determinants that are essential for protective immunity, including the role of the rare and labile cyclic trans-(2,3) pyruvate ketal modification, remain largely unknown. Molecular insights will support the design of synthetic subunit oligosaccharide vaccines. Here, we identified the key antigenic determinants of ST4 CPS with the help of pyruvated and nonpyruvated synthetic repeating unit glycans. Glycan arrays revealed oligosaccharide antigens recognized by antibodies in the human reference serum. Selected depyruvated ST4 oligosaccharides were used to formulate neoglycoconjugates and immunologically evaluated in mice. These oligosaccharides were highly immunogenic, but the resulting antiglycan antibodies showed only limited binding to the natural CPS present on the bacterial surface. Glycan array and surface plasmon resonance analysis of murine polyclonal serum antibodies as well as monoclonal antibodies revealed that terminal sugars are important in directing the immune responses. The pyruvate modification on the oligosaccharide is needed for cross-reactivity with the native CPS. These findings are an important step toward the design of oligosaccharide-based vaccines against S. pneumoniae ST4.
Collapse
Affiliation(s)
- Andreas Geissner
- Department
of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute
for Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Claney L. Pereira
- Department
of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
| | - Melanie Leddermann
- Department
of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
| | - Chakkumkal Anish
- Department
of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
| | - Peter H. Seeberger
- Department
of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute
for Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| |
Collapse
|
45
|
Park HJ, Ko HL, Jung SY, Jo HB, Nam JH. The Characteristics of RNA Vaccine; its Strengths and Weaknesses. ACTA ACUST UNITED AC 2016. [DOI: 10.4167/jbv.2016.46.3.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Hyo-Jung Park
- Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Korea
| | - Hae Li Ko
- Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Korea
| | - Seo-Yeon Jung
- Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Korea
| | - Han-Byeol Jo
- Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Korea
| |
Collapse
|
46
|
Structure and stabilization of the Hendra virus F glycoprotein in its prefusion form. Proc Natl Acad Sci U S A 2015; 113:1056-61. [PMID: 26712026 DOI: 10.1073/pnas.1523303113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hendra virus (HeV) is one of the two prototypical members of the Henipavirus genus of paramyxoviruses, which are designated biosafety level 4 (BSL-4) organisms due to the high mortality rate of Nipah virus (NiV) and HeV in humans. Paramyxovirus cell entry is mediated by the fusion protein, F, in response to binding of a host receptor by the attachment protein. During posttranslational processing, the fusion peptide of F is released and, upon receptor-induced triggering, inserts into the host cell membrane. As F undergoes a dramatic refolding from its prefusion to postfusion conformation, the fusion peptide brings the host and viral membranes together, allowing entry of the viral RNA. Here, we present the crystal structure of the prefusion form of the HeV F ectodomain. The structure shows very high similarity to the structure of prefusion parainfluenza virus 5 (PIV5) F, with the main structural differences in the membrane distal apical loops and the fusion peptide cleavage loop. Functional assays of mutants show that the apical loop can tolerate perturbation in length and surface residues without loss of function, except for residues involved in the stability and conservation of the F protein fold. Structure-based disulfide mutants were designed to anchor the fusion peptide to conformationally invariant residues of the F head. Two mutants were identified that inhibit F-mediated fusion by stabilizing F in its prefusion conformation.
Collapse
|
47
|
McComb RC, Martchenko M. Neutralizing antibody and functional mapping of Bacillus anthracis protective antigen-The first step toward a rationally designed anthrax vaccine. Vaccine 2015; 34:13-9. [PMID: 26611201 DOI: 10.1016/j.vaccine.2015.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/05/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022]
Abstract
Anthrax is defined by the Centers for Disease Control and Prevention as a Category A pathogen for its potential use as a bioweapon. Current prevention treatments include Anthrax Vaccine Adsorbed (AVA). AVA is an undefined formulation of Bacillus anthracis culture supernatant adsorbed to aluminum hydroxide. It has an onerous vaccination schedule, is slow and cumbersome to produce and is slightly reactogenic. Next-generation vaccines are focused on producing recombinant forms of anthrax toxin in a well-defined formulation but these vaccines have been shown to lose potency as they are stored. In addition, studies have shown that a proportion of the antibody response against these vaccines is focused on non-functional, non-neutralizing regions of the anthrax toxin while some essential functional regions are shielded from eliciting an antibody response. Rational vaccinology is a developing field that focuses on designing vaccine antigens based on structural information provided by neutralizing antibody epitope mapping, crystal structure analysis, and functional mapping through amino acid mutations. This information provides an opportunity to design antigens that target only functionally important and conserved regions of a pathogen in order to make a more optimal vaccine product. This review provides an overview of the literature related to functional and neutralizing antibody epitope mapping of the Protective Antigen (PA) component of anthrax toxin.
Collapse
Affiliation(s)
- Ryan C McComb
- Keck Graduate Institute, School of Applied Life Science, 535 Watson Dr., Claremont, CA, United States
| | - Mikhail Martchenko
- Keck Graduate Institute, School of Applied Life Science, 535 Watson Dr., Claremont, CA, United States.
| |
Collapse
|
48
|
Structural and Computational Biology in the Design of Immunogenic Vaccine Antigens. J Immunol Res 2015; 2015:156241. [PMID: 26526043 PMCID: PMC4615220 DOI: 10.1155/2015/156241] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/02/2015] [Indexed: 01/08/2023] Open
Abstract
Vaccination is historically one of the most important medical interventions for the prevention of infectious disease. Previously, vaccines were typically made of rather crude mixtures of inactivated or attenuated causative agents. However, over the last 10–20 years, several important technological and computational advances have enabled major progress in the discovery and design of potently immunogenic recombinant protein vaccine antigens. Here we discuss three key breakthrough approaches that have potentiated structural and computational vaccine design. Firstly, genomic sciences gave birth to the field of reverse vaccinology, which has enabled the rapid computational identification of potential vaccine antigens. Secondly, major advances in structural biology, experimental epitope mapping, and computational epitope prediction have yielded molecular insights into the immunogenic determinants defining protective antigens, enabling their rational optimization. Thirdly, and most recently, computational approaches have been used to convert this wealth of structural and immunological information into the design of improved vaccine antigens. This review aims to illustrate the growing power of combining sequencing, structural and computational approaches, and we discuss how this may drive the design of novel immunogens suitable for future vaccines urgently needed to increase the global prevention of infectious disease.
Collapse
|
49
|
Bennett KM, Gorham RD, Gusti V, Trinh L, Morikis D, Lo DD. Hybrid flagellin as a T cell independent vaccine scaffold. BMC Biotechnol 2015; 15:71. [PMID: 26265529 PMCID: PMC4534063 DOI: 10.1186/s12896-015-0194-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 07/29/2015] [Indexed: 12/12/2022] Open
Abstract
Background To extend the potency of vaccines against infectious diseases, vaccines should be able to exploit multiple arms of the immune system. One component of the immune system that is under-used in vaccine design is the subset of B cells known to be capable of responding to repetitive antigenic epitopes and differentiate into plasma cells even in the absence of T cell help (T-independent, TI). Results To target vaccine responses from T-independent B cells, we reengineered a bacterial Flagellin (FliC) by replacing its exposed D3 domain with a viral envelope protein from Dengue virus (DENV2). The resulting hybrid FliC protein (hFliC) was able to form stable filaments decorated with conformationally intact DENV2 envelope domains. These filaments were not only capable of inducing a T cell-dependent (TD) humoral antibody response, but also significant IgM and IgG3 antibody response in a helper T cell repertoire-restricted transgenic mouse model. Conclusions Our results provide proof-of-principle demonstration that a reengineered hybrid FliC could be used as a platform for polymeric subunit vaccines, enhancing T cell-dependent and possibly inducing T-independent antibody responses from B-1 B cells as well. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0194-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaila M Bennett
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA. .,Bioengineering Interdepartmental Graduate Program, California, USA.
| | - Ronald D Gorham
- Department of Bioengineering, University of California Riverside, California, 92521, USA.
| | - Veronica Gusti
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA.
| | - Lien Trinh
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA.
| | - Dimitrios Morikis
- Department of Bioengineering, University of California Riverside, California, 92521, USA.
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA.
| |
Collapse
|
50
|
Guo N, Duan H, Kachko A, Krause BW, Major ME, Krause PR. Reverse Engineering of Vaccine Antigens Using High Throughput Sequencing-enhanced mRNA Display. EBioMedicine 2015; 2:859-67. [PMID: 26425692 PMCID: PMC4563141 DOI: 10.1016/j.ebiom.2015.06.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 06/22/2015] [Accepted: 06/26/2015] [Indexed: 01/12/2023] Open
Abstract
Vaccine reverse engineering is emerging as an important approach to vaccine antigen identification, recently focusing mainly on structural characterization of interactions between neutralizing monoclonal antibodies (mAbs) and antigens. Using mAbs that bind unknown antigen structures, we sought to probe the intrinsic features of antibody antigen-binding sites with a high complexity peptide library, aiming to identify conformationally optimized mimotope antigens that capture mAb-specific epitopes. Using a high throughput sequencing-enhanced messenger ribonucleic acid (mRNA) display approach, we identified high affinity binding peptides for a hepatitis C virus neutralizing mAb. Immunization with the selected peptides induced neutralizing activity similar to that of the original mAb. Antibodies elicited by the most commonly selected peptides were predominantly against specific epitopes. Thus, using mRNA display to interrogate mAbs permits high resolution identification of functional peptide antigens that direct targeted immune responses, supporting its use in vaccine reverse engineering for pathogens against which potent neutralizing mAbs are available. Research in Context We used a large number of randomly produced small proteins (“peptides”) to identify peptides containing specific protein sequences that bind efficiently to an antibody that can prevent hepatitis C virus infection in cell culture. After the identified peptides were injected into mice, the mice produced their own antibodies with characteristics similar to the original antibody. This approach can provide previously unavailable information about antibody binding and could also be useful in developing new vaccines. mRNA-display/high throughput sequencing identified high affinity peptide binders to monoclonal antibody (mAb). The profile of selected peptides characterized the binding specificity of the selection mAb. Immune responses induced by selected peptides were neutralizing and epitope-focused, mimicking the selection mAb.
Collapse
Affiliation(s)
- Nini Guo
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States
| | - Hongying Duan
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States
| | - Alla Kachko
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States
| | - Benjamin W Krause
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States
| | - Marian E Major
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States
| | - Philip R Krause
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States
| |
Collapse
|