1
|
Waz NT, Milani B, Assoni L, Coelho GR, Sciani JM, Parisotto T, Ferraz LFC, Hakansson AP, Converso TR, Darrieux M. Pneumococcal surface protein A (PspA) prevents killing of Streptococcus pneumoniae by indolicidin. Sci Rep 2024; 14:23517. [PMID: 39384882 PMCID: PMC11464550 DOI: 10.1038/s41598-024-73564-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/18/2024] [Indexed: 10/11/2024] Open
Abstract
Pneumococcal surface protein A (PspA) is an important virulence factor in Streptococcus pneumoniae that binds to lactoferrin and protects the bacterium from the bactericidal action of lactoferricins-cationic peptides released upon lactoferrin proteolysis. The present study investigated if PspA can prevent killing by another cationic peptide, indolicidin. PspA-negative pneumococci were more sensitive to indolicidin-induced killing than bacteria expressing PspA, suggesting that PspA prevents the bactericidal action of indolicidin. Similarly, chemical removal of choline-binding proteins increased sensitivity to indolicidin. The absence of capsule and PspA had an additive effect on pneumococcal killing by the AMP. Furthermore, anti-PspA antibodies enhanced the bactericidal effect of indolicidin on pneumococci, while addition of soluble PspA fragments competitively inhibited indolicidin action. Previous in silico analysis suggests a possible interaction between PspA and indolicidin. Thus, we hypothesize that PspA acts by sequestering indolicidin and preventing it from reaching the bacterial membrane. A specific interaction between PspA and indolicidin was demonstrated by mass spectrometry, confirming that PspA can actively bind to the AMP. These results reinforce the vaccine potential of PspA and suggest a possible mechanism of innate immune evasion employed by pneumococci, which involves binding to cationic peptides and hindering their ability to damage the bacterial membranes.
Collapse
Affiliation(s)
- Natalha T Waz
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Barbara Milani
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lucas Assoni
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | | | - Juliana M Sciani
- Laboratório de Produtos Naturais, Universidade São Francisco, Bragança Paulista, Brazil
| | - Thaís Parisotto
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lucio F C Ferraz
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Anders P Hakansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Lund, Sweden
| | - Thiago R Converso
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Michelle Darrieux
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil.
| |
Collapse
|
2
|
Trentini MM, Rodriguez D, Kanno AI, Goulart C, Darrieux M, de Cerqueira Leite LC. Robust Immune Response and Protection against Lethal Pneumococcal Challenge with a Recombinant BCG-PspA-PdT Prime/Boost Scheme Administered to Neonatal Mice. Vaccines (Basel) 2024; 12:122. [PMID: 38400107 PMCID: PMC10893189 DOI: 10.3390/vaccines12020122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Pneumococcal diseases are an important public health problem, with high mortality rates in young children. Although conjugated pneumococcal vaccines offer high protection against invasive pneumococcal diseases, this is restricted to vaccine serotypes, leading to serotype replacement. Furthermore, the current vaccines do not protect neonates. Therefore, several protein-based pneumococcal vaccines have been studied over the last few decades. Our group established a recombinant BCG expressing rPspA-PdT as a prime/rPspA-PdT boost strategy, which protected adult mice against lethal intranasal pneumococcal challenge. Here, we immunized groups of neonate C57/Bl6 mice (6-10) (at 5 days) with rBCG PspA-PdT and a boost with rPspA-PdT (at 12 days). Controls were saline or each antigen alone. The prime/boost strategy promoted an IgG1 to IgG2c isotype shift compared to protein alone. Furthermore, there was an increase in specific memory cells (T and B lymphocytes) and higher cytokine production (IFN-γ, IL-17, TNF-α, IL-10, and IL-6). Immunization with rBCG PspA-PdT/rPspA-PdT showed 100% protection against pulmonary challenge with the WU2 pneumococcal strain; two doses of rPspA-PdT showed non-significant protection in the neonates. These results demonstrate that a prime/boost strategy using rBCG PspA-PdT/rPspA-PdT is effective in protecting neonates against lethal pneumococcal infection via the induction of strong antibody and cytokine responses.
Collapse
Affiliation(s)
| | - Dunia Rodriguez
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Alex Issamu Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Cibelly Goulart
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Michelle Darrieux
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista 12916-900, Brazil;
| | | |
Collapse
|
3
|
Lane JR, Tata M, Yasmin R, Im H, Briles DE, Orihuela CJ. PspA-mediated aggregation protects Streptococcus pneumoniae against desiccation on fomites. mBio 2023; 14:e0263423. [PMID: 37982608 PMCID: PMC10746202 DOI: 10.1128/mbio.02634-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 11/21/2023] Open
Abstract
IMPORTANCE Spn is a dangerous human pathogen capable of causing pneumonia and invasive disease. The virulence factor PspA has been studied for nearly four decades with well-established roles in pneumococcal evasion of C-reactive protein and neutralization of lactoferricin. Herein, we show that mammalian (m)GAPDH in mucosal secretions promotes aggregation of pneumococci in a PspA-dependent fashion, whereas lactoferrin counters this effect. PspA-mediated GAPDH-dependent bacterial aggregation protected Spn in nasal lavage elutes and grown in vitro from desiccation on fomites. Furthermore, surviving pneumococci within these aggregates retained their ability to colonize naïve hosts after desiccation. We report that Spn binds to and forms protein complexes on its surface composed of PspA, mGAPDH, and lactoferrin. Changes in the levels of these proteins therefore most likely have critical implications on Spn colonization, survival on fomites, and transmission.
Collapse
Affiliation(s)
- Jessica R. Lane
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Muralidhar Tata
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rahena Yasmin
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hansol Im
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David E. Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Carlos J. Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
4
|
Milani B, dos Santos TW, Guerra MES, Oliveira S, Goulart C, André GO, Leite LCC, Converso TR, Darrieux M. Fusion of PspA to detoxified pneumolysin enhances pneumococcal vaccine coverage. PLoS One 2023; 18:e0291203. [PMID: 38096222 PMCID: PMC10721071 DOI: 10.1371/journal.pone.0291203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/23/2023] [Indexed: 12/17/2023] Open
Abstract
Despite the implementation of conjugate vaccines in several countries, S. pneumoniae continues to pose a great burden worldwide, causing around 1 million annual deaths. Pneumococcal proteins have long been investigated as serotype-independent vaccines against this pathogen, with promising results. However, it is a consensus that one antigen alone will not be sufficient to provide long-term protection with wide coverage. Amongst the most well studied pneumococcal proteins are PspA and pneumolysin (Ply), two major virulence factors required by the bacterium for successful invasion of host tissues. PspA is highly immunogenic and protective, but it is structurally variable; pneumolysin is conserved among different pneumococci, but it is toxic to the host. To overcome these limitations, N-terminal PspA fragments have been genetically fused to non-toxic pneumolysin derivatives (PlD) to create PspA_PlD chimeras. Mouse immunization with these fusions confers protection against pneumococcal strains expressing heterologous PspAs, which correlates with antibody-induced complement C3 deposition on the surface of multiple pneumococcal strains. Analysis of mutant strains lacking PspA or Pneumolysin shows that both proteins contribute to the antibody-mediated enhancement in complement deposition induced by the fusion. These results expand previous data evaluating PspA_PlD and demonstrate that the fusion combines the protective traits of both proteins, inducing antibodies that efficiently promote complement deposition on multiple strains and cross-protection.
Collapse
Affiliation(s)
- Barbara Milani
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Tanila Wood dos Santos
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
- Programa de Pós-Graduação Interunidades em Biotecnologia-USP-IPT-IB, São Paulo, Brazil
| | | | - Sheila Oliveira
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Cibelly Goulart
- Centro de Biotecnologia, Instituto Butantan, São Paulo, Brazil
| | - Greiciely O. André
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | | | - Thiago R Converso
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Michelle Darrieux
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| |
Collapse
|
5
|
Shafaghi M, Bahadori Z, Barzi SM, Afshari E, Madanchi H, Mousavi SF, Shabani AA. A new candidate epitope-based vaccine against PspA PhtD of Streptococcus pneumoniae: a computational experimental approach. Front Cell Infect Microbiol 2023; 13:1271143. [PMID: 38035337 PMCID: PMC10684780 DOI: 10.3389/fcimb.2023.1271143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Pneumococcus is an important respiratory pathogen that is associated with high rates of death in newborn children and the elderly. Given the disadvantages of current polysaccharide-based vaccines, the most promising alternative for developing improved vaccines may be to use protein antigens with different roles in pneumococcus virulence. PspA and PhtD, highly immunogenic surface proteins expressed by almost all pneumococcal strains, are capable of eliciting protective immunity against lethal infections. Methods In this study using immunoinformatics approaches, we constructed one fusion construct (called PAD) by fusing the immunodominant regions of PspA from families 1 & 2 (PA) to the immunodominant regions of PhtD (PD). The objective of this project was to test the immunogenicity of the fusion protein PAD and to compare its protective activity against S. pneumoniae infection with PA or PD alone and a combination of PA and PD. The prediction of physicochemical properties, antigenicity, allergenicity, toxicity, and 3D-structure of the constructs, as well as molecular docking with HLA receptor and immune simulation were performed using computational tools. Finally, mice were immunized and the serum levels of antibodies/cytokines and functionality of antibodies in vitro were evaluated after immunization. The mice survival rates and decrease of bacterial loads in the blood/spleen were examined following the challenge. Results The computational analyses indicated the proposed constructs could be antigenic, non-allergenic, non-toxic, soluble and able to elicit robust immune responses. The results of actual animal experiments revealed the candidate vaccines could induce the mice to produce high levels of antibodies and cytokines. The complement-mediated bactericidal activity of antibodies was confirmed and the antibodies provided favorable survival in immunized mice after bacterial challenge. In general, the experimental results verified the immunoinformatics studies. Conclusion For the first time this report presents novel peptide-based vaccine candidates consisting of immunodominant regions of PspA and PhtD antigens. The obtained findings confirmed that the fusion formulation could be relatively more efficient than the individual and combination formulations. The results propose that the fusion protein alone could be used as a serotype-independent pneumococcal vaccine or as an effective partner protein for a conjugate polysaccharide vaccine.
Collapse
Affiliation(s)
- Mona Shafaghi
- Department of Medical Biotechnology, faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Zohreh Bahadori
- Department of Medical Biotechnology, faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Elnaz Afshari
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Madanchi
- Department of Medical Biotechnology, faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Ali Akbar Shabani
- Department of Medical Biotechnology, faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
6
|
Lane JR, Tata M, Yasmin R, Im H, Briles DE, Orihuela CJ. PspA-mediated aggregation protects Streptococcus pneumoniae against desiccation on fomites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559802. [PMID: 37808718 PMCID: PMC10557681 DOI: 10.1101/2023.09.27.559802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Streptococcus pneumoniae (Spn) resides in the nasopharynx where it can disseminate to cause disease. One key Spn virulence factor is pneumococcal surface protein A (PspA), which promotes survival by blocking the antimicrobial peptide lactoferricin. PspA has also been shown to mediate attachment to dying epithelial cells in the lower airway due to its binding of cell surface-bound mammalian (m)GAPDH. Importantly, the role of PspA during colonization is not well understood. Wildtype Spn was present in nasal lavage elutes collected from asymptomatically colonized mice at levels ~10-fold higher that its isogenic PspA-deficient mutant (ΔpspA). Wildtype Spn also formed aggregates in mucosal secretions composed of sloughed epithelial cells and hundreds of pneumococci, whereas ΔpspA did not. Spn within the center of these aggregates better survived prolonged desiccation on fomites than individual pneumococci and were capable of infecting naïve mice, indicating PspA-mediated aggregation conferred a survival/transmission advantage. Incubation of Spn in saline containing mGAPDH also enhanced tolerance to desiccation, but only for wildtype Spn. mGAPDH was sufficient to cause low-level aggregation of wildtype Spn but not ΔpspA. In strain WU2, the subdomain of PspA responsible for binding GAPDH (aa230-281) is ensconced within the lactoferrin (LF)-binding domain (aa167-288). We observed that LF inhibited GAPDH-mediated aggregation and desiccation tolerance. Using surface plasmon resonance, we determined that Spn forms multimeric complexes of PspA-GAPDH-LF on its surface and that LF dislodges GAPDH. Our findings have important implications regarding pneumococcal colonization/transmission processes and ongoing PspA-focused immunization efforts for this deadly pathogen.
Collapse
Affiliation(s)
- Jessica R. Lane
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35209, United States
| | - Muralidhar Tata
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35209, United States
| | - Rahena Yasmin
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35209, United States
| | - Hansol Im
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35209, United States
| | - David E. Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35209, United States
| | - Carlos J. Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35209, United States
| |
Collapse
|
7
|
Carneiro GB, Castro JT, Davi M, Miyaji EN, Ladant D, Oliveira MLS. Immune responses and protection against Streptococcus pneumoniae elicited by recombinant Bordetella pertussis adenylate cyclase (CyaA) carrying fragments of pneumococcal surface protein A, PspA. Vaccine 2023:S0264-410X(23)00570-4. [PMID: 37236818 DOI: 10.1016/j.vaccine.2023.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
Streptococcus pneumoniae is a common agent of important human diseases such as otitis media, pneumonia, meningitis and sepsis. Current available vaccines that target capsular polysaccharides induce protection against invasive disease and nasopharyngeal colonization in children, yet their efficacy is limited to the serotypes included in the formulations. The virulence factor Pneumococcal Surface Protein A (PspA) interacts with host immune system and helps the bacteria to evade phagocytosis. Due to its essential role in virulence, PspA is an important vaccine candidate. Here we have tested a delivery system based on the adenylate cyclase toxin of Bordetella pertussis (CyaA) to induce immune responses against PspA in mice. CyaA was engineered to express fragments of the N-terminal region of PspAs from clades 2 and 4 (A2 and A4) and the resulting proteins were used in immunization experiments in mice. The recombinant CyaA-A2 and CyaA-A4 proteins were able to induce high levels of anti-PspA antibodies that reacted with pneumococcal strains expressing either PspA2 or PspA4. Moreover, reactivity of the antibodies against pneumococcal strains that express PspAs from clades 3 and 5 (PspA3 and PspA5) was also observed. A formulation containing CyaA-A2 and CyaA-A4 was able to protect mice against invasive pneumococcal challenges with isolates that express PspA2, PspA4 or PspA5. Moreover, a CyaA-A2-A4 fusion protein induced antibodies at similar levels and with similar reactivity as the formulation containing both proteins, and protected mice against the invasive challenge. Our results indicate that CyaA-PspA proteins are good candidates to induce broad protection against pneumococcal isolates.
Collapse
Affiliation(s)
| | | | - Marilyne Davi
- Institut Pasteur, Université de Paris Cité, CNRS UMR 3528, Unité de Biochimie des Interactions Macromoléculaires, Paris, France
| | | | - Daniel Ladant
- Institut Pasteur, Université de Paris Cité, CNRS UMR 3528, Unité de Biochimie des Interactions Macromoléculaires, Paris, France.
| | | |
Collapse
|
8
|
Shafaghi M, Bahadori Z, Madanchi H, Ranjbar MM, Shabani AA, Mousavi SF. Immunoinformatics-aided design of a new multi-epitope vaccine adjuvanted with domain 4 of pneumolysin against Streptococcus pneumoniae strains. BMC Bioinformatics 2023; 24:67. [PMID: 36829109 PMCID: PMC9951839 DOI: 10.1186/s12859-023-05175-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Streptococcus pneumoniae (Pneumococcus) has remained a leading cause of fatal infections such as pneumonia, meningitis, and sepsis. Moreover, this pathogen plays a major role in bacterial co-infection in patients with life-threatening respiratory virus diseases such as influenza and COVID-19. High morbidity and mortality in over one million cases, especially in very young children and the elderly, are the main motivations for pneumococcal vaccine development. Due to the limitations of the currently marketed polysaccharide-based vaccines, non-serotype-specific protein-based vaccines have received wide research interest in recent years. One step further is to identify high antigenic regions within multiple highly-conserved proteins in order to develop peptide vaccines that can affect various stages of pneumococcal infection, providing broader serotype coverage and more effective protection. In this study, immunoinformatics tools were used to design an effective multi-epitope vaccine in order to elicit neutralizing antibodies against multiple strains of pneumococcus. RESULTS The B- and T-cell epitopes from highly protective antigens PspA (clades 1-5) and PhtD were predicted and immunodominant peptides were linked to each other with proper linkers. The domain 4 of Ply, as a potential TLR4 agonist adjuvant candidate, was attached to the end of the construct to enhance the immunogenicity of the epitope vaccine. The evaluation of the physicochemical and immunological properties showed that the final construct was stable, soluble, antigenic, and non-allergenic. Furthermore, the protein was found to be acidic and hydrophilic in nature. The protein 3D-structure was built and refined, and the Ramachandran plot, ProSA-web, ERRAT, and Verify3D validated the quality of the final model. Molecular docking analysis showed that the designed construct via Ply domain 4 had a strong interaction with TLR4. The structural stability of the docked complex was confirmed by molecular dynamics. Finally, codon optimization was performed for gene expression in E. coli, followed by in silico cloning in the pET28a(+) vector. CONCLUSION The computational analysis of the construct showed acceptable results, however, the suggested vaccine needs to be experimentally verified in laboratory to ensure its safety and immunogenicity.
Collapse
Affiliation(s)
- Mona Shafaghi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Research Center of Biotechnology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Zohreh Bahadori
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Research Center of Biotechnology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Hamid Madanchi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Research Center of Biotechnology, Semnan University of Medical Sciences, Semnan, Iran
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Mehdi Ranjbar
- Agricultural Research, Education, and Extension Organization (AREEO), Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Ali Akbar Shabani
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Research Center of Biotechnology, Semnan University of Medical Sciences, Semnan, Iran.
| | | |
Collapse
|
9
|
Lane JR, Tata M, Briles DE, Orihuela CJ. A Jack of All Trades: The Role of Pneumococcal Surface Protein A in the Pathogenesis of Streptococcus pneumoniae. Front Cell Infect Microbiol 2022; 12:826264. [PMID: 35186799 PMCID: PMC8847780 DOI: 10.3389/fcimb.2022.826264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Streptococcus pneumoniae (Spn), or the pneumococcus, is a Gram-positive bacterium that colonizes the upper airway. Spn is an opportunistic pathogen capable of life-threatening disease should it become established in the lungs, gain access to the bloodstream, or disseminate to vital organs including the central nervous system. Spn is encapsulated, allowing it to avoid phagocytosis, and current preventative measures against infection include polyvalent vaccines composed of capsular polysaccharide corresponding to its most prevalent serotypes. The pneumococcus also has a plethora of surface components that allow the bacteria to adhere to host cells, facilitate the evasion of the immune system, and obtain vital nutrients; one family of these are the choline-binding proteins (CBPs). Pneumococcal surface protein A (PspA) is one of the most abundant CBPs and confers protection against the host by inhibiting recognition by C-reactive protein and neutralizing the antimicrobial peptide lactoferricin. Recently our group has identified two new roles for PspA: binding to dying host cells via host-cell bound glyceraldehyde 3-phosphate dehydrogenase and co-opting of host lactate dehydrogenase to enhance lactate availability. These properties have been shown to influence Spn localization and enhance virulence in the lower airway, respectively. Herein, we review the impact of CBPs, and in particular PspA, on pneumococcal pathogenesis. We discuss the potential and limitations of using PspA as a conserved vaccine antigen in a conjugate vaccine formulation. PspA is a vital component of the pneumococcal virulence arsenal - therefore, understanding the molecular aspects of this protein is essential in understanding pneumococcal pathogenesis and utilizing PspA as a target for treating or preventing pneumococcal pneumonia.
Collapse
Affiliation(s)
| | | | | | - Carlos J. Orihuela
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
10
|
Luck JN, Tettelin H, Orihuela CJ. Sugar-Coated Killer: Serotype 3 Pneumococcal Disease. Front Cell Infect Microbiol 2020; 10:613287. [PMID: 33425786 PMCID: PMC7786310 DOI: 10.3389/fcimb.2020.613287] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Capsular polysaccharide (CPS), which surrounds the bacteria, is one of the most significant and multifaceted contributors to Streptococcus pneumoniae virulence. Capsule prevents entrapment in mucus during colonization, traps water to protect against desiccation, can serve as an energy reserve, and protects the bacterium against complement-mediated opsonization and immune cell phagocytosis. To date, 100 biochemically and serologically distinct capsule types have been identified for S. pneumoniae; 20 to 30 of which have well-defined propensity to cause opportunistic human infection. Among these, serotype 3 is perhaps the most problematic as serotype 3 infections are characterized as having severe clinical manifestations including empyema, bacteremia, cardiotoxicity, and meningitis; consequently, with a fatality rate of 30%-47%. Moreover, serotype 3 resists antibody-mediated clearance despite its inclusion in the current 13-valent conjugate vaccine formulation. This review covers the role of capsule in pneumococcal pathogenesis and the importance of serotype 3 on human disease. We discuss how serotype 3 capsule synthesis and presentation on the bacterial surface is distinct from other serotypes, the biochemical and physiological properties of this capsule type that facilitate its ability to cause disease, and why existing vaccines are unable to confer protection. We conclude with discussion of the clonal properties of serotype 3 and how these have changed since introduction of the 13-valent vaccine in 2000.
Collapse
Affiliation(s)
- Jennifer N. Luck
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carlos J. Orihuela
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
11
|
The Modified Surface Killing Assay Distinguishes between Protective and Nonprotective Antibodies to PspA. mSphere 2019; 4:4/6/e00589-19. [PMID: 31826968 PMCID: PMC6908419 DOI: 10.1128/msphere.00589-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The most important finding of this study is that the MSKA can be used as an in vitro functional assay. Such an assay will be critical for the development of PspA-containing vaccines. The other important findings relate to the locations and nature of the protection-eliciting epitopes of PspA. There are limited prior data on the locations of protection-eliciting PspA epitopes, but those data along with the data presented here make it clear that there is not a single epitope or domain of PspA that can elicit protective antibody and there exists at least one region of the αHD which seldom elicits protective antibody. Moreover, these data, in concert with prior data, strongly make the case that protective epitopes in the αHD are highly conformational (≥100-amino-acid fragments of the αHD are required), whereas at least some protection-eliciting epitopes in the proline-rich domain are encoded by ≤15-amino-acid sequences. Pneumococcal surface protein A (PspA) elicits antibody protective against lethal challenge by Streptococcus pneumoniae and is a candidate noncapsular antigen for inclusion in vaccines. Evaluation of immunity to PspA in human trials would be greatly facilitated by an in vitro functional assay able to distinguish protective from nonprotective antibodies to PspA. Mouse monoclonal antibodies (MAbs) to PspA can mediate killing by human granulocytes in the modified surface killing assay (MSKA). To determine if the MSKA can distinguish between protective and nonprotective MAbs, we examined seven MAbs to PspA. All bound recombinant PspA, as detected by enzyme-linked immunosorbent assay and Western blotting; four gave strong passive protection against fatal challenge, two were nonprotective, and the seventh one only delayed death. The four that were able to provide strong passive protection were also most able to enhance killing in the MSKA, the two that were not protective in mice were not effective in the MSKA, and the MAb that was only weakly protective in mice was weakly effective in the MSKA (P < 0.001). One of the four most protective MAbs tested reacted to the proline-rich domain of PspA. Two of the other most protective MAbs and the weakly protective MAb reacted with a fragment from PspA’s α-helical domain (αHD), containing amino acids (aa) 148 to 247 from the N terminus of PspA. The fourth highly protective MAb recognized none of the overlapping 81- or 100-aa fragments of PspA. The two nonprotective MAbs recognized a more N-terminal αHD fragment (aa 48 to 147). IMPORTANCE The most important finding of this study is that the MSKA can be used as an in vitro functional assay. Such an assay will be critical for the development of PspA-containing vaccines. The other important findings relate to the locations and nature of the protection-eliciting epitopes of PspA. There are limited prior data on the locations of protection-eliciting PspA epitopes, but those data along with the data presented here make it clear that there is not a single epitope or domain of PspA that can elicit protective antibody and there exists at least one region of the αHD which seldom elicits protective antibody. Moreover, these data, in concert with prior data, strongly make the case that protective epitopes in the αHD are highly conformational (≥100-amino-acid fragments of the αHD are required), whereas at least some protection-eliciting epitopes in the proline-rich domain are encoded by ≤15-amino-acid sequences.
Collapse
|
12
|
Briles DE, Paton JC, Mukerji R, Swiatlo E, Crain MJ. Pneumococcal Vaccines. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0028-2018. [PMID: 31858954 PMCID: PMC10921951 DOI: 10.1128/microbiolspec.gpp3-0028-2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Indexed: 01/14/2023] Open
Abstract
Streptococcus pneumoniae is a Gram-Positive pathogen that is a major causative agent of pneumonia, otitis media, sepsis and meningitis across the world. The World Health Organization estimates that globally over 500,000 children are killed each year by this pathogen. Vaccines offer the best protection against S. pneumoniae infections. The current polysaccharide conjugate vaccines have been very effective in reducing rates of invasive pneumococcal disease caused by vaccine type strains. However, the effectiveness of these vaccines have been somewhat diminished by the increasing numbers of cases of invasive disease caused by non-vaccine type strains, a phenomenon known as serotype replacement. Since, there are currently at least 98 known serotypes of S. pneumoniae, it may become cumbersome and expensive to add many additional serotypes to the current 13-valent vaccine, to circumvent the effect of serotype replacement. Hence, alternative serotype independent strategies, such as vaccination with highly cross-reactive pneumococcal protein antigens, should continue to be investigated to address this problem. This chapter provides a comprehensive discussion of pneumococcal vaccines past and present, protein antigens that are currently under investigation as vaccine candidates, and other alternatives, such as the pneumococcal whole cell vaccine, that may be successful in reducing current rates of disease caused by S. pneumoniae.
Collapse
Affiliation(s)
- D E Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - J C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, 5005, Australia
| | - R Mukerji
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - E Swiatlo
- Section of Infectious Diseases, Southeast Louisiana Veterans Health Care System, New Orleans, LA
| | - M J Crain
- Department of Pediatrics and Microbiology, University of Alabama at Birmingham
| |
Collapse
|
13
|
Akbari E, Negahdari B, Faraji F, Behdani M, Kazemi-Lomedasht F, Habibi-Anbouhi M. Protective responses of an engineered PspA recombinant antigen against Streptococcus pneumoniae. ACTA ACUST UNITED AC 2019; 24:e00385. [PMID: 31763198 PMCID: PMC6864353 DOI: 10.1016/j.btre.2019.e00385] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/20/2019] [Accepted: 10/08/2019] [Indexed: 11/24/2022]
Abstract
In this study, two immunogenic antigens based on recombinant PspA proteins were immunized mice. The protective effects of developed anti-PspA antibodies in mice in intranasal and intraperitoneal challenges were proved. Based on the obtained results, immunization with the B-regions of PspA antigens are crucial in protection of challenged mice with S. pneumoniae strains.
Streptococcus pneumoniae is a major pathogen in human respiratory tract which causes significant morbidity and mortality across from the world. Currently available vaccines are not completely effective and cannot cover all pathogenic strains so there is an important need to develop an alternative cost-effective vaccine, based on conserved protein antigens. Pneumococcal surface protein A (PspA) is one of interesting candidates for development of a serotype-independent vaccine against pneumococcal infections. PspA is grouped into two major families with five clades, and broad-reacting PspA-based vaccines should contain at least one functional fragment from each of the two families. In this study, we developed two immunogenic antigens based on recombinant PspA proteins that including the different antigenic regions of PspA from both two families. The cross-reactivity of antibodies elicited against two PspA proteins PspAB1-5 and PspA4ABC and their role in complement deposition with three strains of pneumococci were tested. The protective effects of developed anti-PspA antibodies in mice in intranasal and intraperitoneal challenges were evaluated using a strain from clade 2. Sera from immunized mice with PspAB1-5 in comparison with PspA4ABC was able to deposit more C3 complement component on surface of pneumococci bearing diverse PspA from both families 1 and 2, and immunized mice with the PspAB1-5 showed a higher protection than PspA4ABC in pneumococcal challenges. The obtained results from this study indicate that a PspA-based antigen composed of B region from all clades in addition to conserved domains, can provide a significant protection against multiple strains of S. pneumoniae and may overcome the limitation of polysaccharide vaccines.
Collapse
Affiliation(s)
- Elaheh Akbari
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faraji
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | | |
Collapse
|
14
|
Lagousi T, Basdeki P, Routsias J, Spoulou V. Novel Protein-Based Pneumococcal Vaccines: Assessing the Use of Distinct Protein Fragments Instead of Full-Length Proteins as Vaccine Antigens. Vaccines (Basel) 2019; 7:vaccines7010009. [PMID: 30669439 PMCID: PMC6466302 DOI: 10.3390/vaccines7010009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Non-serotype-specific protein-based pneumococcal vaccines have received extensive research focus due to the limitations of polysaccharide-based vaccines. Pneumococcal proteins (PnPs), universally expressed among serotypes, may induce broader immune responses, stimulating humoral and cellular immunity, while being easier to manufacture and less expensive. Such an approach has raised issues mainly associated with sequence/level of expression variability, chemical instability, as well as possible undesirable reactogenicity and autoimmune properties. A step forward employs the identification of highly-conserved antigenic regions within PnPs with the potential to retain the benefits of protein antigens. Besides, their low-cost and stable construction facilitates the combination of several antigenic regions or peptides that may impair different stages of pneumococcal disease offering even wider serotype coverage and more efficient protection. This review discusses the up-to-date progress on PnPs that are currently under clinical evaluation and the challenges for their licensure. Focus is given on the progress on the identification of antigenic regions/peptides within PnPs and their evaluation as vaccine candidates, accessing their potential to overcome the issues associated with full-length protein antigens. Particular mention is given of the use of newer delivery system technologies including conjugation to Toll-like receptors (TLRs) and reformulation into nanoparticles to enhance the poor immunogenicity of such antigens.
Collapse
Affiliation(s)
- Theano Lagousi
- First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Immunobiology Research Laboratory and Infectious Diseases Department "MAKKA," Athens Medical School, 11527 Athens, Greece.
| | - Paraskevi Basdeki
- First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Immunobiology Research Laboratory and Infectious Diseases Department "MAKKA," Athens Medical School, 11527 Athens, Greece.
| | - John Routsias
- Department of Microbiology, Athens Medical School, 11527 Athens, Greece.
| | - Vana Spoulou
- First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Immunobiology Research Laboratory and Infectious Diseases Department "MAKKA," Athens Medical School, 11527 Athens, Greece.
| |
Collapse
|
15
|
Mukerji R, Hendrickson C, Genschmer KR, Park SS, Bouchet V, Goldstein R, Lefkowitz EJ, Briles DE. The diversity of the proline-rich domain of pneumococcal surface protein A (PspA): Potential relevance to a broad-spectrum vaccine. Vaccine 2018; 36:6834-6843. [PMID: 30293761 DOI: 10.1016/j.vaccine.2018.08.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/18/2018] [Accepted: 08/19/2018] [Indexed: 01/28/2023]
Abstract
Pneumococcal surface protein A (PspA) is a surface exposed, highly immunogenic protein of Streptococcus pneumoniae. Its N-terminal α-helical domain (αHD) elicits protective antibody in humans and animals that can protect mice from fatal infections with pneumococci and can be detected in vitro with opsonophagocytosis assays. The proline-rich domain (PRD) in the center of the PspA sequence can also elicit protection. This study revealed that although the sequence of PRD was diverse, PRD from different pneumococcal isolates contained many shared elements. The inferred amino acid sequences of 123 such PRDs, which were analyzed by assembly and alignment-free (AAF) approaches, formed three PRD groups. Of these sequences, 45 were classified as Group 1, 19 were classified as Group 2, and 59 were classified as Group 3. All Group 3 sequences contained a highly conserved 22-amino acid non-proline block (NPB). A significant polymorphism was observed, however, at a single amino acid position within NPB. Each of the three PRD groups had characteristic patterns of short amino acid repeats, with most of the repeats being found in more than one PRD group. One of these repeats, PKPEQP as well as the NPB were previously shown to elicit protective antibodies in mice. In this study, we found that sera from 12 healthy human adult volunteers contained antibodies to all three PRD groups. This suggested that a PspA-containing vaccine containing carefully selected PRDs and αHDs could redundantly cover the known diversity of PspA. Such an approach might reduce the chances of PspA variants escaping a PspA vaccine's immunity.
Collapse
Affiliation(s)
- Reshmi Mukerji
- Department of Microbiology, University of Alabama at Birmingham, United States
| | - Curtis Hendrickson
- Center for Clinical and Translational Sciences, University of Alabama at Birmingham, United States
| | - Kristopher R Genschmer
- Department of Microbiology, University of Alabama at Birmingham, United States; Department of Medicine, Division of Pulmonary, Allergy and Critical Care, United States
| | - Sang-Sang Park
- Department of Microbiology, University of Alabama at Birmingham, United States
| | - Valérie Bouchet
- Section of Molecular Genetics, Maxwell Finland Laboratory for Infectious Diseases, Division of Pediatric Infectious Diseases, Boston University Medical Center, Boston, MA 02118, United States
| | - Richard Goldstein
- Section of Molecular Genetics, Maxwell Finland Laboratory for Infectious Diseases, Division of Pediatric Infectious Diseases, Boston University Medical Center, Boston, MA 02118, United States
| | - Elliot J Lefkowitz
- Department of Microbiology, University of Alabama at Birmingham, United States; Center for Clinical and Translational Sciences, University of Alabama at Birmingham, United States
| | - David E Briles
- Department of Microbiology, University of Alabama at Birmingham, United States.
| |
Collapse
|
16
|
A pneumococcal vaccine combination with two proteins containing PspA families 1 and 2 can potentially protect against a wide range of Streptococcus pneumoniae strains. Immunol Res 2018; 66:528-536. [PMID: 30128745 DOI: 10.1007/s12026-018-9016-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
17
|
Comparison of Immunogenicity and Protection of Two Pneumococcal Protein Vaccines Based on PsaA and PspA. Infect Immun 2018; 86:IAI.00916-17. [PMID: 29610257 DOI: 10.1128/iai.00916-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/22/2018] [Indexed: 12/11/2022] Open
Abstract
Streptococcus pneumoniae is a major cause of invasive pneumococcal disease, septicemia, and meningitis that can result in high morbidity rates in children under 5 years old. The current polysaccharide-based vaccines can provide type-specific immunity, but a broad-spectrum vaccine would provide greater coverage. Therefore, developing pneumococcal-protein-based vaccines that can extend to more serum types is highly important. In this study, we vaccinated mice via the subcutaneous (s.c.) route with a systemic vaccine that is a mixture of fusion protein PsaA-PspA23 and a single protein, PspA4, with aluminum hydroxide as an adjuvant. As a comparison, mice were immunized intranasally with a mucosal vaccine that is a mixture of PspA2-PA-BLP (where PA is protein anchor and BLP is bacterium-like particle) and PspA4-PA-BLP, via the intranasal (i.n.) route. The two immunization processes were followed by challenge with Streptococcus pneumoniae bacteria from two different PspA families. Specific IgG titers in the serum and specific IgA titers in the mucosa were determined following immunizations. Bacterial loads and survival rates after challenge were compared. Both the systemic vaccine and the mucosal vaccine induced a significant increase of IgG against PspAs. Only the mucosal vaccine also induced specific IgA in the mucosa. The two vaccines provided protection, but each vaccine showed an advantage. The systemic vaccine induced higher levels of serum antibodies, whereas the mucosal vaccine limited the bacterial load in the lung and blood. Therefore, coimmunizations with the two types of vaccines may be implemented in the future.
Collapse
|
18
|
Yun KW, Choi EH, Lee HJ. Genetic diversity of pneumococcal surface protein A in invasive pneumococcal isolates from Korean children, 1991-2016. PLoS One 2017; 12:e0183968. [PMID: 29131872 PMCID: PMC5683564 DOI: 10.1371/journal.pone.0183968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 08/15/2017] [Indexed: 12/02/2022] Open
Abstract
Pneumococcal surface protein A (PspA) is an important virulence factor of pneumococci and has been investigated as a primary component of a capsular serotype-independent pneumococcal vaccine. Thus, we sought to determine the genetic diversity of PspA to explore its potential as a vaccine candidate. Among the 190 invasive pneumococcal isolates collected from Korean children between 1991 and 2016, two (1.1%) isolates were found to have no pspA by multiple polymerase chain reactions. The full length pspA genes from 185 pneumococcal isolates were sequenced. The length of pspA varied, ranging from 1,719 to 2,301 base pairs with 55.7–100% nucleotide identity. Based on the sequences of the clade-defining regions, 68.7% and 49.7% were in PspA family 2 and clade 3/family 2, respectively. PspA clade types were correlated with genotypes using multilocus sequence typing and divided into several subclades based on diversity analysis of the N-terminal α-helical regions, which showed nucleotide sequence identities of 45.7–100% and amino acid sequence identities of 23.1–100%. Putative antigenicity plots were also diverse among individual clades and subclades. The differences in antigenicity patterns were concentrated within the N-terminal 120 amino acids. In conclusion, the N-terminal α-helical domain, which is known to be the major immunogenic portion of PspA, is genetically variable and should be further evaluated for antigenic differences and cross-reactivity between various PspA types from pneumococcal isolates.
Collapse
Affiliation(s)
- Ki Wook Yun
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | - Eun Hwa Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | - Hoan Jong Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| |
Collapse
|
19
|
Recombinant BCG expressing a PspA-PdT fusion protein protects mice against pneumococcal lethal challenge in a prime-boost strategy. Vaccine 2017; 35:1683-1691. [PMID: 28242071 DOI: 10.1016/j.vaccine.2017.02.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/13/2017] [Accepted: 02/14/2017] [Indexed: 11/24/2022]
Abstract
Pneumococcal proteins have been evaluated as genetically-conserved potential vaccine candidates. We have previously demonstrated that a fragment of PspA in fusion with PdT (rPspA-PdT) induced protective immune responses in mice. However, purified proteins have shown poor immunogenicity and often require the combination with strong adjuvants and booster doses. Here, we investigated the use of a Bacillus Calmette-Guérin (BCG) strain, a well-established prophylactic vaccine for tuberculosis with known adjuvant properties, for delivery of the PspA-PdT fusion protein. Immunization of mice in a prime-boost strategy, using rPspA-PdT as a boost, demonstrated that rBCG PspA-PdT/rPspA-PdT was able to induce an antibody response against both proteins, promoting an IgG1 to IgG2 antibody isotype shift. Sera from rBCG PspA-PdT/rPspA-PdT immunized mice showed antibodies able to bind to the pneumococcal surface and promoted higher complement deposition when compared with WT-BCG/rPspA-PdT or a single dose of rPspA-PdT. In addition, these antisera inhibited the cytolytic activity of Ply. Production of interleukin-6 (IL-6), gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α) was increased in splenocytes culture. Furthermore, a higher expression of CD69 early activation molecule was observed on splenic CD4+ T cells from mice immunized with rBCG PspA-PdT before and after the protein booster dose. Finally, immunization with rBCG PspA-PdT/rPspA-PdT protected mice against pneumococcal lethal challenge. These results support the further investigation of recombinant BCG strains to express pneumococcal proteins, which could be administered in early stages of life and lead to protective pneumococcal immunity in infants and children.
Collapse
|
20
|
Mirza S, Benjamin WH, Coan PA, Hwang SA, Winslett AK, Yother J, Hollingshead SK, Fujihashi K, Briles DE. The effects of differences in pspA alleles and capsular types on the resistance of Streptococcus pneumoniae to killing by apolactoferrin. Microb Pathog 2016; 99:209-219. [PMID: 27569531 DOI: 10.1016/j.micpath.2016.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/15/2016] [Accepted: 08/23/2016] [Indexed: 11/18/2022]
Abstract
Pneumococcal surface protein A (PspA) is the only pneumococcal surface protein known to strongly bind lactoferrin on the bacterial surface. In the absence of PspA Streptococcus pneumoniae becomes more susceptible to killing by human apolactoferrin (apo-hLf), the iron-free form of lactoferrin. In the present study we examined diverse strains of S. pneumoniae that differed by 2 logs in their susceptibility to apo-hLf. Among these strains, the amount of apo-hLf that bound to cell surface PspA correlated directly with the resistance of the strain to killing by apo-hLf. Moreover examination of different pspA alleles on shared genetic backgrounds revealed that those PspAs that bound more lactoferrin conferred greater resistance to killing by apo-hLf. The effects of capsule on killing of pneumococci by apo-hLf were generally small, but on one genetic background, however, the lack of capsule was associated with 4-times as much apo-hLf binding and 30-times more resistance to killing by apo-hLf. Overall these finding strongly support the hypothesis that most of the variation in the ability of apo-hLf is dependent on the variation in the binding of apo-hLf to surface PspA and this binding is dependent on variation in PspA as well as variation in capsule which may enhance killing by reducing the binding of apo-hLf to PspA.
Collapse
Affiliation(s)
- Shaper Mirza
- Department of Biology, Lahore University of Management Sciences, Pakistan; Division of Epidemiology, Human Genetics and Environmental Health, School of Public Health, University of Texas, Health Science Center, Brownsville Regional Campus, TX, USA.
| | - William H Benjamin
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Patricia A Coan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Shen-An Hwang
- Department of Pathology and Laboratory Medicine, Medical School University of Texas Health Science Center, Houston, TX, USA
| | - Anne-Kathryn Winslett
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Janet Yother
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Susan K Hollingshead
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kohtaro Fujihashi
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Department of Pediatrics Dentistry, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David E Briles
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
21
|
Li Y, Beitelshees M, Fang L, Hill A, Ahmadi MK, Chen M, Davidson BA, Knight P, Smith RJ, Andreadis ST, Hakansson AP, Jones CH, Pfeifer BA. In situ pneumococcal vaccine production and delivery through a hybrid biological-biomaterial vector. SCIENCE ADVANCES 2016; 2:e1600264. [PMID: 27419235 PMCID: PMC4942325 DOI: 10.1126/sciadv.1600264] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/06/2016] [Indexed: 05/14/2023]
Abstract
The type and potency of an immune response provoked during vaccination will determine ultimate success in disease prevention. The basis for this response will be the design and implementation of antigen presentation to the immune system. Whereas direct antigen administration will elicit some form of immunological response, a more sophisticated approach would couple the antigen of interest to a vector capable of broad delivery formats and designed for heightened response. New antigens associated with pneumococcal disease virulence were used to test the delivery and adjuvant capabilities of a hybrid biological-biomaterial vector consisting of a bacterial core electrostatically coated with a cationic polymer. The hybrid design provides (i) passive and active targeting of antigen-presenting cells, (ii) natural and multicomponent adjuvant properties, (iii) dual intracellular delivery mechanisms, and (iv) a simple formulation mechanism. In addition, the hybrid format enables device-specific, or in situ, antigen production and consolidation via localization within the bacterial component of the vector. This capability eliminates the need for dedicated antigen production and purification before vaccination efforts while leveraging the aforementioned features of the overall delivery device. We present the first disease-specific utilization of the vector toward pneumococcal disease highlighted by improved immune responses and protective capabilities when tested against traditional vaccine formulations and a range of clinically relevant Streptococcus pneumoniae strains. More broadly, the results point to similar levels of success with other diseases that would benefit from the production, delivery, and efficacy capabilities offered by the hybrid vector.
Collapse
Affiliation(s)
- Yi Li
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Marie Beitelshees
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Lei Fang
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Andrew Hill
- Abcombi Biosciences Inc., Buffalo, NY 14260–4200, USA
| | - Mahmoud Kamal Ahmadi
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Mingfu Chen
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Bruce A. Davidson
- Department of Anesthesiology, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Paul Knight
- Department of Anesthesiology, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Randall J. Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
- Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
| | - Anders P. Hakansson
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
- Division of Experimental Infection Medicine, Department of Laboratory Medicine, Lund University, Malmö SE-20502, Sweden
| | - Charles H. Jones
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
- Abcombi Biosciences Inc., Buffalo, NY 14260–4200, USA
- Corresponding author. (C.H.J.); (B.A.P.)
| | - Blaine A. Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260–4200, USA
- Abcombi Biosciences Inc., Buffalo, NY 14260–4200, USA
- Corresponding author. (C.H.J.); (B.A.P.)
| |
Collapse
|
22
|
Li Y, Hill A, Beitelshees M, Shao S, Lovell JF, Davidson BA, Knight PR, Hakansson AP, Pfeifer BA, Jones CH. Directed vaccination against pneumococcal disease. Proc Natl Acad Sci U S A 2016; 113:6898-903. [PMID: 27274071 PMCID: PMC4922154 DOI: 10.1073/pnas.1603007113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Immunization strategies against commensal bacterial pathogens have long focused on eradicating asymptomatic carriage as well as disease, resulting in changes in the colonizing microflora with unknown future consequences. Additionally, current vaccines are not easily adaptable to sequence diversity and immune evasion. Here, we present a "smart" vaccine that leverages our current understanding of disease transition from bacterial carriage to infection with the pneumococcus serving as a model organism. Using conserved surface proteins highly expressed during virulent transition, the vaccine mounts an immune response specifically against disease-causing bacterial populations without affecting carriage. Aided by a delivery technology capable of multivalent surface display, which can be adapted easily to a changing clinical picture, results include complete protection against the development of pneumonia and sepsis during animal challenge experiments with multiple, highly variable, and clinically relevant pneumococcal isolates. The approach thus offers a unique and dynamic treatment option readily adaptable to other commensal pathogens.
Collapse
Affiliation(s)
- Yi Li
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260
| | | | - Marie Beitelshees
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260
| | - Shuai Shao
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260
| | - Bruce A Davidson
- Department of Anesthesiology, University at Buffalo, The State University of New York, Buffalo, NY 14260
| | - Paul R Knight
- Department of Anesthesiology, University at Buffalo, The State University of New York, Buffalo, NY 14260; Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260
| | - Anders P Hakansson
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260; Division of Experimental Infection Medicine, Department of Laboratory Medicine, Lund University, SE-221 00 Malmo, Sweden
| | - Blaine A Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260; Abcombi Biosciences Inc., Buffalo, NY 14260;
| | | |
Collapse
|
23
|
Khan MN, Pichichero ME. The host immune dynamics of pneumococcal colonization: implications for novel vaccine development. Hum Vaccin Immunother 2015; 10:3688-99. [PMID: 25668673 DOI: 10.4161/21645515.2014.979631] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The human nasopharynx (NP) microbiota is complex and diverse and Streptococcus pneumoniae (pneumococcus) is a frequent member. In the first few years of life, children experience maturation of their immune system thereby conferring homeostatic balance in which pneumococci are typically rendered as harmless colonizers in the upper respiratory environment. Pneumococcal carriage declines in many children before they acquire capsular-specific antibodies, suggesting a capsule antibody-independent mechanism of natural protection against pneumococcal carriage in early childhood. A child's immune system in the first few years of life is Th2-skewed so as to avoid inflammation-induced immunopathology. Understanding Th1/Th2 and Th17 ontogeny in early life and how adjuvant vaccine formulations shift the balance of T helper-cell differentiation, may facilitate the development of new protein-based pneumococcal vaccines. This article will discuss the immune dynamics of pneumococcal colonization in infants. The discussion aims to benefit the design and improvement of protein subunit-based next-generation pneumococcal vaccines.
Collapse
Affiliation(s)
- M Nadeem Khan
- a Center for Infectious Diseases and Immunology; Rochester General Hospital Research Institute ; Rochester , NY USA
| | | |
Collapse
|
24
|
Combined effects of lactoferrin and lysozyme on Streptococcus pneumoniae killing. Microb Pathog 2015; 89:7-17. [PMID: 26298002 DOI: 10.1016/j.micpath.2015.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 08/13/2015] [Accepted: 08/18/2015] [Indexed: 11/20/2022]
Abstract
Streptococcus pneumoniae is a common colonizer of the human nasopharynx, which can occasionally spread to sterile sites, causing diseases such as otitis media, sinusitis, pneumonia, meningitis and bacteremia. Human apolactoferrin (ALF) and lysozyme (LZ) are two important components of the mucosal innate immune system, exhibiting lytic effects against a wide range of microorganisms. Since they are found in similar niches of the host, it has been proposed that ALF and LZ could act synergistically in controlling bacterial spread throughout the mucosa. The combination of ALF and LZ has been shown to enhance killing of different pathogens in vitro, with ALF facilitating the latter action of LZ. The aim of the present work was to investigate the combined effects of ALF and LZ on S pneumoniae. Concomitant addition of ALF and LZ had a synergistic killing effect on one of the pneumococci tested. Furthermore, the combination of ALF and ALZ was more bactericidal than lysozyme alone in all pneumococcal strains. Pneumococcal surface protein A (PspA), an important vaccine candidate, partially protects pneumococci from ALF mediated killing, while antibodies against one PspA enhance killing of the homologous strain by ALF. However, the serological variability of this molecule could limit the effect of anti-PspA antibodies on different pneumococci. Therefore, we investigated the ability of anti-PspA antibodies to increase ALF-mediated killing of strains that express different PspAs, and found that antisera to the N-terminal region of PspA were able to increase pneumococcal lysis by ALF, independently of the sequence similarities between the molecule expressed on the bacterial surface and that used to produce the antibodies. LF binding to the pneumococcal surface was confirmed by flow cytometry, and found to be inhibited in presence of anti-PspA antibodies. On a whole, the results suggest a contribution of ALF and LZ to pneumococcal clearance, and confirm PspA's ability to interact with ALF.
Collapse
|
25
|
Lu J, Sun T, Wang D, Dong Y, Xu M, Hou H, Kong FT, Liang C, Gu T, Chen P, Sun S, Lv X, Jiang C, Kong W, Wu Y. Protective Immune Responses Elicited by Fusion Protein Containing PsaA and PspA Fragments. Immunol Invest 2015; 44:482-96. [DOI: 10.3109/08820139.2015.1037956] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
26
|
Lin H, Peng Y, Lin Z, Zhang S, Guo Y. Development of a conjugate vaccine against invasive pneumococcal disease based on capsular polysaccharides coupled with PspA/family 1 protein of Streptococcus pneumoniae. Microb Pathog 2015; 83-84:35-40. [DOI: 10.1016/j.micpath.2015.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 04/28/2015] [Accepted: 04/28/2015] [Indexed: 11/30/2022]
|
27
|
Kuipers K, Daleke-Schermerhorn MH, Jong WSP, ten Hagen-Jongman CM, van Opzeeland F, Simonetti E, Luirink J, de Jonge MI. Salmonella outer membrane vesicles displaying high densities of pneumococcal antigen at the surface offer protection against colonization. Vaccine 2015; 33:2022-9. [PMID: 25776921 DOI: 10.1016/j.vaccine.2015.03.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/24/2015] [Accepted: 03/04/2015] [Indexed: 11/17/2022]
Abstract
Bacterial outer membrane vesicles (OMVs) are attractive vaccine formulations because they have intrinsic immunostimulatory properties. In principle, heterologous antigens incorporated into OMVs will elicit specific immune responses, especially if presented at the vesicle surface and thus optimally exposed to the immune system. In this study, we explored the feasibility of our recently developed autotransporter Hbp platform, designed to efficiently and simultaneously display multiple antigens at the surface of bacterial OMVs, for vaccine development. Using two Streptococcus pneumoniae proteins as model antigens, we showed that intranasally administered Salmonella OMVs displaying high levels of antigens at the surface induced strong protection in a murine model of pneumococcal colonization, without the need for a mucosal adjuvant. Importantly, reduction in bacterial recovery from the nasal cavity was correlated with local production of antigen-specific IL-17A. Furthermore, the protective efficacy and the production of antigen-specific IL-17A, and local and systemic IgGs, were all improved at increased concentrations of the displayed antigen. This discovery highlights the importance of an adequate antigen expression system for development of recombinant OMV vaccines. In conclusion, our findings demonstrate the suitability of the Hbp platform for development of a new generation of OMV vaccines, and illustrate the potential of using this approach to develop a broadly protective mucosal pneumococcal vaccine.
Collapse
Affiliation(s)
- Kirsten Kuipers
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria H Daleke-Schermerhorn
- Section Molecular Microbiology, Department of Molecular Cell Biology, Faculty of Earth and Life Sciences, VU University, Amsterdam, The Netherlands; Abera Bioscience AB, Stockholm, Sweden
| | - Wouter S P Jong
- Section Molecular Microbiology, Department of Molecular Cell Biology, Faculty of Earth and Life Sciences, VU University, Amsterdam, The Netherlands; Abera Bioscience AB, Stockholm, Sweden
| | - Corinne M ten Hagen-Jongman
- Section Molecular Microbiology, Department of Molecular Cell Biology, Faculty of Earth and Life Sciences, VU University, Amsterdam, The Netherlands; Abera Bioscience AB, Stockholm, Sweden
| | - Fred van Opzeeland
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elles Simonetti
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joen Luirink
- Section Molecular Microbiology, Department of Molecular Cell Biology, Faculty of Earth and Life Sciences, VU University, Amsterdam, The Netherlands; Abera Bioscience AB, Stockholm, Sweden.
| | - Marien I de Jonge
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Correlation between in vitro complement deposition and passive mouse protection of anti-pneumococcal surface protein A monoclonal antibodies. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 22:99-107. [PMID: 25410204 DOI: 10.1128/cvi.00001-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The shortcomings of the licensed polysaccharide-based pneumococcal vaccine are driving efforts toward development of a protein-based vaccine that is serotype independent and effective in all age groups. An opsonophagocytic killing assay (OPKA) is used to evaluate the antibody response against polysaccharide-based pneumococcal vaccines. However, the OPKA is not reliable for noncapsular antigens. Thus, there is a need to develop an in vitro surrogate for protection for protein vaccine candidates like pneumococcal surface antigen A (PspA). PspA is a serologically variable cell surface virulence factor. Based on its sequence, PspA has been classified into families 1 (clade 1 and 2), 2 (clades 3, 4 and 5), and 3 (clade 6). Here, we report the characterization of 18 IgG anti-PspA monoclonal antibodies (anti-PspA(hkR36A) MAbs) generated from mice immunized with heat-killed strain R36A (clade 2). An enzyme-linked immunosorbent assay (ELISA)-based analysis of the reactivity of the MAbs with recombinant PspAs from the 6 clades indicated that they were family 1 specific. This was confirmed by flow cytometry using a hyperimmune serum generated against PspA from R36A. Eight MAbs that bind at least one clade 1- and clade 2-expressing strain were evaluated for complement deposition, bactericidal activity, and passive protection. The anti-PspA(hkR36A) MAb-dependent deposition of complement on pneumococci showed a positive correlation with passive protection against strain WU2 (r = 0.8783, P = 0.0041). All of our protective MAbs showed bactericidal activity; however, not all MAbs that exhibited bactericidal activity conferred protection in vivo. The protective MAbs described here can be used to identify conserved protection eliciting B cell epitopes for engineering a superior PspA-based vaccine.
Collapse
|
29
|
Pertussis toxin improves immune responses to a combined pneumococcal antigen and leads to enhanced protection against Streptococcus pneumoniae. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:972-81. [PMID: 24807055 DOI: 10.1128/cvi.00134-14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pneumococcal surface protein A (PspA) is a candidate antigen for the composition of protein-based vaccines against Streptococcus pneumoniae. While searching for efficient adjuvants for PspA-based vaccines, our group has described the potential of combining PspA with the whole-cell pertussis vaccine (wP). When given to mice through the nasal route, a formulation composed of PspA from clade 5 (PspA5) and wP (PspA5-wP) induced high levels of antibodies and protection against challenges with different pneumococcal strains. PspA5-wP also induced the secretion of interleukin 17 (IL-17) by splenocytes and the infiltration of leukocytes in the lungs after challenge. Here, we show that protection against a pneumococcal invasive challenge was completely abrogated in μMT(-/-) mice, which are deficient in the maturation of B cells, illustrating the importance of antibodies in the survival elicited by the PspA5-wP vaccine. Moreover, passive immunization showed that IgG purified from the sera of mice immunized with PspA5-wP conferred significant protection to naive mice, whereas the respective F(ab')2 did not. Additionally, in vivo depletion of complement abolished protection against the pneumococcal challenge. The combination of PspA5 with wild-type or mutant Bordetella pertussis strains or with purified components showed that the pertussis toxin (PT)-containing formulations induced the highest levels of antibodies and protection. This suggests that the adjuvant activity of wP in the PspA5 model is mediated at least in part by PT. The sera from mice immunized with such formulations displayed high IgG binding and induction of complement deposition on the pneumococcal surface in vitro, which is consistent with the in vivo results.
Collapse
|
30
|
Mapping of epitopes recognized by antibodies induced by immunization of mice with PspA and PspC. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:940-8. [PMID: 24807052 DOI: 10.1128/cvi.00239-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pneumococcal surface protein A (PspA) and pneumococcal surface protein C (PspC) are important candidates for an alternative vaccine against pneumococcal infections. Since these antigens show variability, the use of variants that do not afford broad protection may lead to the selection of vaccine escape bacteria. Epitopes capable of inducing antibodies with broad cross-reactivities should thus be the preferred antigens. In this work, experiments using peptide arrays show that most linear epitopes recognized by antibodies induced in mice against different PspAs were located at the initial 44 amino acids of the mature protein and that antibodies against these linear epitopes did not confer protection against a lethal challenge. Conversely, linear epitopes recognized by antibodies to PspC included the consensus sequences involved in the interaction with human factor H and secretory immunoglobulin A (sIgA). Since linear epitopes of PspA were not protective, larger overlapping fragments containing 100 amino acids of PspA of strain Rx1 were constructed (fragments 1 to 7, numbered from the N terminus) to permit the mapping of antibodies with conformational epitopes not represented in the peptide arrays. Antibodies from mice immunized with fragments 1, 2, 4, and 5 were capable of binding onto the surface of pneumococci and mediating protection against a lethal challenge. The fact that immunization of mice with 100-amino-acid fragments located at the more conserved N-terminal region of PspA (fragments 1 and 2) induced protection against a pneumococcal challenge indicates that the induction of antibodies against conformational epitopes present at this region may be important in strategies for inducing broad protection against pneumococci.
Collapse
|
31
|
Identification of glycosylated regions in pneumococcal PspA conjugated to serotype 6B capsular polysaccharide. Glycoconj J 2014; 31:259-69. [DOI: 10.1007/s10719-014-9519-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/26/2014] [Accepted: 03/03/2014] [Indexed: 10/25/2022]
|
32
|
Kolberg J, Aase A, Naess LM, Aaberge IS, Caugant DA. Human antibody responses to pneumococcal surface protein A and capsular polysaccharides during acute and convalescent stages of invasive disease in adult patients. Pathog Dis 2013; 70:40-50. [DOI: 10.1111/2049-632x.12106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 09/13/2013] [Accepted: 09/23/2013] [Indexed: 11/30/2022] Open
Affiliation(s)
- Jan Kolberg
- Department of Bacteriology and Immunology; Norwegian Institute of Public Health; Oslo Norway
| | - Audun Aase
- Department of Bacteriology and Immunology; Norwegian Institute of Public Health; Oslo Norway
| | - Lisbeth M. Naess
- Department of Bacteriology and Immunology; Norwegian Institute of Public Health; Oslo Norway
| | - Ingeborg S. Aaberge
- Department of Bacteriology and Immunology; Norwegian Institute of Public Health; Oslo Norway
| | - Dominique A. Caugant
- Department of Bacteriology and Immunology; Norwegian Institute of Public Health; Oslo Norway
| |
Collapse
|
33
|
Haughney SL, Petersen LK, Schoofs AD, Ramer-Tait AE, King JD, Briles DE, Wannemuehler MJ, Narasimhan B. Retention of structure, antigenicity, and biological function of pneumococcal surface protein A (PspA) released from polyanhydride nanoparticles. Acta Biomater 2013; 9:8262-71. [PMID: 23774257 DOI: 10.1016/j.actbio.2013.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 02/03/2023]
Abstract
Pneumococcal surface protein A (PspA) is a choline-binding protein which is a virulence factor found on the surface of all Streptococcus pneumoniae strains. Vaccination with PspA has been shown to be protective against a lethal challenge with S. pneumoniae, making it a promising immunogen for use in vaccines. Herein the design of a PspA-based subunit vaccine using polyanhydride nanoparticles as a delivery platform is described. Nanoparticles based on sebacic acid (SA), 1,6-bis-(p-carboxyphenoxy)hexane (CPH) and 1,8-bis-(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG), specifically 50:50 CPTEG:CPH and 20:80 CPH:SA, were used to encapsulate and release PspA. The protein released from the nanoparticle formulations retained its primary and secondary structure as well as its antigenicity. The released PspA was also biologically functional based on its ability to bind to apolactoferrin and prevent its bactericidal activity against Escherichia coli. When the PspA nanoparticle formulations were administered subcutaneously to mice they elicited a high titer and high avidity anti-PspA antibody response. Together these studies provide a framework for the rational design of a vaccine against S. pneumoniae based on polyanhydride nanoparticles.
Collapse
|
34
|
Darrieux M, Goulart C, Briles D, Leite LCDC. Current status and perspectives on protein-based pneumococcal vaccines. Crit Rev Microbiol 2013; 41:190-200. [DOI: 10.3109/1040841x.2013.813902] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Characterization of protective immune responses induced by pneumococcal surface protein A in fusion with pneumolysin derivatives. PLoS One 2013; 8:e59605. [PMID: 23533636 PMCID: PMC3606166 DOI: 10.1371/journal.pone.0059605] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 02/15/2013] [Indexed: 01/09/2023] Open
Abstract
Pneumococcal surface protein A (PspA) and Pneumolysin derivatives (Pds) are important vaccine candidates, which can confer protection in different models of pneumococcal infection. Furthermore, the combination of these two proteins was able to increase protection against pneumococcal sepsis in mice. The present study investigated the potential of hybrid proteins generated by genetic fusion of PspA fragments to Pds to increase cross-protection against fatal pneumococcal infection. Pneumolisoids were fused to the N-terminus of clade 1 or clade 2 pspA gene fragments. Mouse immunization with the fusion proteins induced high levels of antibodies against PspA and Pds, able to bind to intact pneumococci expressing a homologous PspA with the same intensity as antibodies to rPspA alone or the co-administered proteins. However, when antibody binding to pneumococci with heterologous PspAs was examined, antisera to the PspA-Pds fusion molecules showed stronger antibody binding and C3 deposition than antisera to co-administered proteins. In agreement with these results, antisera against the hybrid proteins were more effective in promoting the phagocytosis of bacteria bearing heterologous PspAs in vitro, leading to a significant reduction in the number of bacteria when compared to co-administered proteins. The respective antisera were also capable of neutralizing the lytic activity of Pneumolysin on sheep red blood cells. Finally, mice immunized with fusion proteins were protected against fatal challenge with pneumococcal strains expressing heterologous PspAs. Taken together, the results suggest that PspA-Pd fusion proteins comprise a promising vaccine strategy, able to increase the immune response mediated by cross-reactive antibodies and complement deposition to heterologous strains, and to confer protection against fatal challenge.
Collapse
|
36
|
Hotomi M, Togawa A, Kono M, Ikeda Y, Takei S, Hollingshead SK, Briles DE, Suzuki K, Yamanaka N. PspA family distribution, antimicrobial resistance and serotype of Streptococcus pneumoniae isolated from upper respiratory tract infections in Japan. PLoS One 2013; 8:e58124. [PMID: 23483982 PMCID: PMC3590126 DOI: 10.1371/journal.pone.0058124] [Citation(s) in RCA: 18] [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: 09/15/2012] [Accepted: 01/30/2013] [Indexed: 11/19/2022] Open
Abstract
Background The protection against pneumococcal infections provided by currently available pneumococcal polysaccharide conjugate vaccines are restricted to the limited number of the serotypes included in the vaccine. In the present study, we evaluated the distribution of the pneumococcal capsular type and surface protein A (PspA) family of pneumococcal isolates from upper respiratory tract infections in Japan. Methods A total of 251 S. pneumoniae isolates from patients seeking treatment for upper respiratory tract infections were characterized for PspA family, antibiotic resistance and capsular type. Results Among the 251 pneumococci studied, the majority (49.4%) was identified as belonging to PspA family 2, while most of the remaining isolates (44.6%) belonged to family 1. There were no significant differences between the distributions of PspA1 versus PspA2 isolates based on the age or gender of the patient, source of the isolates or the isolates’ susceptibilities to penicillin G. In contrast, the frequency of the mefA gene presence and of serotypes 15B and 19F were statistically more common among PspA2 strains. Conclusion The vast majority of pneumococci isolated from the middle ear fluids, nasal discharges/sinus aspirates or pharyngeal secretions represented PspA families 1 and 2. Capsular serotypes were generally not exclusively associated with certain PspA families, although some capsular types showed a much higher proportion of either PspA1 or PspA2. A PspA-containing vaccine would potentially provide high coverage against pneumococcal infectious diseases because it would be cross-protective versus invasive disease with the majority of pneumococci infecting children and adults.
Collapse
Affiliation(s)
- Muneki Hotomi
- Department of Otolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama-shi, Wakayama, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mukerji R, Mirza S, Roche AM, Widener RW, Croney CM, Rhee DK, Weiser JN, Szalai AJ, Briles DE. Pneumococcal surface protein A inhibits complement deposition on the pneumococcal surface by competing with the binding of C-reactive protein to cell-surface phosphocholine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:5327-35. [PMID: 23105137 PMCID: PMC3517878 DOI: 10.4049/jimmunol.1201967] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In the presence of normal serum, complement component C3 is deposited on pneumococci primarily via the classical pathway. Pneumococcal surface protein A (PspA), a major virulence factor of pneumococci, effectively inhibits C3 deposition. PspA's C terminus has a choline-binding domain that anchors PspA to the phosphocholine (PC) moieties on the pneumococcal surface. C-reactive protein (CRP), another important host defense molecule, also binds to PC, and CRP binding to pneumococci enhances complement C3 deposition through the classical pathway. Using flow cytometry of PspA(+) and PspA(-) strains, we observed that the absence of PspA led to exposure of PC, enhanced the surface binding of CRP, and increased the deposition of C3. Moreover, when the PspA(-) mutant was incubated with a pneumococcal eluate containing native PspA, there was decreased deposition of CRP and C3 on the pneumococcal surface compared with incubation with an eluate from a PspA(-) strain. This inhibition was not observed when a recombinant PspA fragment, which lacks the choline-binding region of PspA, was added to the PspA(-) mutant. Also, there was much greater C3 deposition onto the PspA(-) pneumococcus when exposed to normal mouse serum from wild-type mice as compared with that from CRP knockout mice. Furthermore, when CRP knockout mouse serum was replenished with CRP, there was a dose-dependent increase in C3 deposition. The combined data reveal a novel mechanism of complement inhibition by a bacterial protein: inhibition of CRP surface binding and, thus, diminution of CRP-mediated complement deposition.
Collapse
Affiliation(s)
- Reshmi Mukerji
- Department of Microbiology, University of Alabama at Birmingham
| | - Shaper Mirza
- University of Texas School of Public Health Division of Epidemiology Brownsville regional campus Brownsville TX
| | - Aoife M. Roche
- Department of Microbiology, School of Medicine, University of Pennsylvania
| | | | | | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Jeffrey N. Weiser
- Department of Microbiology, School of Medicine, University of Pennsylvania
| | - Alexander J. Szalai
- Department of Microbiology, University of Alabama at Birmingham
- Division of Immunology Department of Medicine, University of Alabama at Birmingham
| | - David E. Briles
- Department of Microbiology, University of Alabama at Birmingham
- Department of Pediatrics, University of Alabama at Birmingham
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
38
|
Vadesilho CFM, Ferreira DM, Moreno AT, Chavez-Olortegui C, Machado de Avila RA, Oliveira MLS, Ho PL, Miyaji EN. Characterization of the antibody response elicited by immunization with pneumococcal surface protein A (PspA) as recombinant protein or DNA vaccine and analysis of protection against an intranasal lethal challenge with Streptococcus pneumoniae. Microb Pathog 2012; 53:243-9. [PMID: 22981893 DOI: 10.1016/j.micpath.2012.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 08/27/2012] [Accepted: 08/29/2012] [Indexed: 11/28/2022]
Abstract
Pneumococcal surface protein A (PspA) is an important candidate for a vaccine against pneumococcal infections. DNA vaccines expressing PspA were shown to protect mice against intraperitoneal and colonization challenge models in mice. We now show that a DNA vaccine expressing PspA from clade 4 (pSec-pspA4Pro) is also able to elicit protection against an intranasal lethal challenge model at levels similar to the recombinant protein PspA4Pro adjuvanted with alum. PspA4Pro + alum induced an IgG response characterized by a high IgG1/IgG2a ratio, leading to a lack of binding of anti-PspA IgG2a antibodies to intact pneumococci in vitro, which is in contrast to the response elicited by pSec-pspA4Pro. Epitopes recognized by the sera were mapped and antibodies induced by immunization with PspA4Pro + alum showed positive reaction with several synthetic peptides, mostly located in the first half of the protein. On the other hand, antibodies induced by the DNA vaccine showed reactivity with only two peptides. Though both strategies were protective against the intranasal lethal challenge model, the elicited humoral responses differ significantly, with the detection of important differences in the Fc (IgG1/IgG2a ratios) and Fab (recognized epitopes) regions of the induced antibodies.
Collapse
Affiliation(s)
- Cintia F M Vadesilho
- Centro de Biotecnologia, Instituto Butantan, Av Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hakansson AP, Roche-Hakansson H, Mossberg AK, Svanborg C. Apoptosis-like death in bacteria induced by HAMLET, a human milk lipid-protein complex. PLoS One 2011; 6:e17717. [PMID: 21423701 PMCID: PMC3053380 DOI: 10.1371/journal.pone.0017717] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 02/11/2011] [Indexed: 11/20/2022] Open
Abstract
Background Apoptosis is the primary means for eliminating unwanted cells in multicellular organisms in order to preserve tissue homeostasis and function. It is characterized by distinct changes in the morphology of the dying cell that are orchestrated by a series of discrete biochemical events. Although there is evidence of primitive forms of programmed cell death also in prokaryotes, no information is available to suggest that prokaryotic death displays mechanistic similarities to the highly regulated programmed death of eukaryotic cells. In this study we compared the characteristics of tumor and bacterial cell death induced by HAMLET, a human milk complex of alpha-lactalbumin and oleic acid. Methodology/Principal Findings We show that HAMLET-treated bacteria undergo cell death with mechanistic and morphologic similarities to apoptotic death of tumor cells. In Jurkat cells and Streptococcus pneumoniae death was accompanied by apoptosis-like morphology such as cell shrinkage, DNA condensation, and DNA degradation into high molecular weight fragments of similar sizes, detected by field inverse gel electrophoresis. HAMLET was internalized into tumor cells and associated with mitochondria, causing a rapid depolarization of the mitochondrial membrane and bound to and induced depolarization of the pneumococcal membrane with similar kinetic and magnitude as in mitochondria. Membrane depolarization in both systems required calcium transport, and both tumor cells and bacteria were found to require serine protease activity (but not caspase activity) to execute cell death. Conclusions/Significance Our results suggest that many of the morphological changes and biochemical responses associated with apoptosis are present in prokaryotes. Identifying the mechanisms of bacterial cell death has the potential to reveal novel targets for future antimicrobial therapy and to further our understanding of core activation mechanisms of cell death in eukaryote cells.
Collapse
Affiliation(s)
- Anders P Hakansson
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York, United States of America.
| | | | | | | |
Collapse
|
40
|
The proline-rich region of pneumococcal surface proteins A and C contains surface-accessible epitopes common to all pneumococci and elicits antibody-mediated protection against sepsis. Infect Immun 2010; 78:2163-72. [PMID: 20194601 DOI: 10.1128/iai.01199-09] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pneumococcal surface protein A (PspA) and PspC of Streptococcus pneumoniae are surface virulence proteins that interfere with complement deposition and elicit protective immune responses. The C-terminal halves of PspA and PspC have some structural similarity and contain highly cross-reactive proline-rich (PR) regions. In many PR regions of PspA and PspC, there exists an almost invariant nonproline block (NPB) of about 33 amino acids. Neither the PR regions nor their NPB exhibit the alpha-helical structure characteristic of much of the protection-eliciting N-terminal portions of PspA and PspC. Prior studies of PspA and PspC as immunogens focused primarily on the alpha-helical regions of these molecules that lack the PR and NPB regions. This report shows that immunization with recombinant PR (rPR) molecules and passive immunization with monoclonal antibodies reactive with either NPB or PR epitopes are protective against infection in mice. PR regions of both PspA and PspC were antibody accessible on the pneumococcal surface. Our results indicate that while PspA could serve as a target of these protective antibodies in invasive infections, PspC might not. When antibody responses to rPR immunogens were evaluated by using flow cytometry to measure antibody binding to live pneumococci, it was observed that the mice that survived subsequent challenge produced significantly higher levels of antibodies reactive with exposed PR epitopes than the mice that became moribund. Due to their conservation and cross-reactivity, the PR regions and NPB regions represent potential vaccine targets capable of eliciting cross-protection immunity against pneumococcal infection.
Collapse
|
41
|
Immunization of mice with single PspA fragments induces antibodies capable of mediating complement deposition on different pneumococcal strains and cross-protection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:439-46. [PMID: 20089795 DOI: 10.1128/cvi.00430-09] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PspA is an important candidate for a vaccine with serotype-independent immunity against pneumococcal infections. Based on sequence relatedness, PspA has been classified into three families comprising six clades. We have previously addressed the cross-reactivity of antibodies against PspA fragments containing the N-terminal and proline-rich regions of PspA from clades 1 to 5 (PspA1, PspA2, PspA3, PspA4, and PspA5) by Western blot analysis and reported that anti-PspA4 and anti-PspA5 were able to recognize pneumococci expressing PspA proteins from all of the clades analyzed. We have now analyzed the functional capacity of these antibodies to bind and to mediate complement deposition on intact bacteria in vitro. Our results show that both PspA4 and PspA5 elicit antibodies that are able to bind and to mediate complement deposition efficiently on pneumococcal strains bearing PspA proteins from clades 1 to 5. Moreover, mice immunized with PspA4 and PspA5 were protected against an intranasal lethal challenge with strains expressing PspA proteins from the two major families. PspA4 and PspA5 are thus able to induce antibodies with a high degree of cross-reactivity in vitro, which is reflected in cross-protection of mice. We have also analyzed the contribution of the nonproline (NonPro) block within the conserved proline-rich region to the reactivity of anti-PspA antibodies, and the results indicate that N-terminal alpha-helical region, the blocks of proline repeats, and the NonPro region can influence the degree of cross-reactivity of antibodies to PspA.
Collapse
|
42
|
PspA family fusion proteins delivered by attenuated Salmonella enterica serovar Typhimurium extend and enhance protection against Streptococcus pneumoniae. Infect Immun 2009; 77:4518-28. [PMID: 19687204 DOI: 10.1128/iai.00486-09] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Pneumococcal surface protein A (PspA) is highly immunogenic and can induce a protective immune response against pneumococcal infection. PspA is divided into two major families based on serological variability: family 1 and family 2. To provide broad protection, PspA proteins from pneumococcal strains Rx1 (family 1) and EF5668 (family 2) were combined to form two PspA fusion proteins, PspA/Rx1-EF5668 and PspA/EF5668-Rx1. Each protein was fused to a type II secretion signal and delivered by a recombinant attenuated Salmonella vaccine (RASV). Both PspA/Rx1-EF5668 and PspA/EF5668-Rx1 were synthesized in the RASV and secreted into the periplasm and supernatant. The fusion proteins reacted strongly with both anti-PspA/Rx1 and anti-PspA/EF5668 antisera. Oral immunization of BALB/c mice with RASV synthesizing either PspA fusion protein elicited serum immunoglobulin G (IgG) and mucosal IgA responses against both families of PspA. Analysis of IgG isotypes (IgG2a and IgG1) indicated a strong Th1 bias to the immune responses to both proteins. Sera from mice immunized with RASV synthesizing PspA/Rx1-EF5668 bound to the surface and directed C3 complement deposition on representative strains from all five PspA clades. Immunization with RASV synthesizing either protein protected mice against intraperitoneal challenge with Streptococcus pneumoniae WU2 strain (family 1), intravenous challenge with S. pneumoniae 3JYP2670 strain (family 2), and intranasal challenge with S. pneumoniae A66.1 (family 1). The PspA/Rx1-EF5668 protein elicited significantly greater protection than PspA/EF5668-Rx1, PspA/Rx1, or PspA/EF5668. These results indicate an RASV synthesizing a PspA fusion protein representing both PspA families constitutes an effective antipneumococcal vaccine, extending and enhancing protection against multiple strains of S. pneumoniae.
Collapse
|
43
|
Rohatgi S, Dutta D, Tahir S, Sehgal D. Molecular Dissection of Antibody Responses against Pneumococcal Surface Protein A: Evidence for Diverse DH-Less Heavy Chain Gene Usage and Avidity Maturation. THE JOURNAL OF IMMUNOLOGY 2009; 182:5570-85. [PMID: 19380805 DOI: 10.4049/jimmunol.0803254] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/biosynthesis
- Antibodies, Bacterial/genetics
- Antibodies, Bacterial/metabolism
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/metabolism
- Antibody Affinity/genetics
- Antibody Diversity/genetics
- Bacterial Proteins/immunology
- Base Sequence
- Epitopes, B-Lymphocyte/metabolism
- Female
- Gene Deletion
- Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics
- Hybridomas
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/metabolism
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/metabolism
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Molecular Sequence Data
- Multigene Family/immunology
- Streptococcus pneumoniae/immunology
Collapse
Affiliation(s)
- Soma Rohatgi
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | | | | | |
Collapse
|
44
|
Tai SS. Streptococcus pneumoniaeProtein Vaccine Candidates: Properties, Activities and Animal Studies. Crit Rev Microbiol 2008; 32:139-53. [PMID: 16893751 DOI: 10.1080/10408410600822942] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Streptococcus pneumoniae is a causative agent for community acquired pneumonia, bacteremia, acute otitis media, and meningitis. Recent emergence of multi-drug resistant clinical isolates prompts the need of effective vaccine for the prevention of disease. The licensed polysaccharide-based pneumococcal vaccines only elicit protective antibodies against the infection of serotypes that are included in the vaccine. To broaden the protection, the use of pneumococcal proteins will be a feasible and preferable alternative. This communication provides a review on the biochemical properties of these protein candidates, their immunization results in animal studies, and perspectives on the development of protein-based pneumococcal vaccine.
Collapse
Affiliation(s)
- Stanley S Tai
- Department of Microbiology, College of Medicine, Howard University, Washington, DC 20059, USA.
| |
Collapse
|
45
|
Lin H, Lin Z, Wang Y, Guo Y. Expression and immunogenicity of PspA family 1 of Streptococcus pneumoniae 6B. J Biotechnol 2008. [DOI: 10.1016/j.jbiotec.2008.07.439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
46
|
Darrieux M, Moreno AT, Ferreira DM, Pimenta FC, de Andrade ALSS, Lopes APY, Leite LCC, Miyaji EN. Recognition of pneumococcal isolates by antisera raised against PspA fragments from different clades. J Med Microbiol 2008; 57:273-278. [DOI: 10.1099/jmm.0.47661-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pneumococcal surface protein A (PspA) is an important vaccine candidate against pneumococcal infections, capable of inducing protection in different animal models. Based on its structural diversity, it has been suggested that a PspA-based vaccine should contain at least one fragment from each of the two major families (family 1, comprising clades 1 and 2, and family 2, comprising clades 3, 4 and 5) in order to elicit broad protection. This study analysed the recognition of a panel of 35 pneumococcal isolates bearing different PspAs by antisera raised against the N-terminal regions of PspA clades 1 to 5. The antiserum to PspA clade 4 was found to show the broadest cross-reactivity, being able to recognize pneumococcal strains containing PspAs of all clades in both families. The cross-reactivity of antibodies elicited against a PspA hybrid including the N-terminal region of clade 1 fused to a shorter and more divergent fragment (clade-defining region, or CDR) of clade 4 (PspA1–4) was also tested, and revealed a strong recognition of isolates containing clades 1, 4 and 5, and weaker reactions with clades 2 and 3. The analysis of serum reactivity against different PspA regions further revealed that the complete N-terminal region rather than just the CDR should be included in an anti-pneumococcal vaccine. A PspA-based vaccine is thus proposed to be composed of the whole N-terminal region of clades 1 and 4, which could also be expressed as a hybrid protein.
Collapse
Affiliation(s)
- Michelle Darrieux
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil, 1500, 05509-900, São Paulo, SP, Brazil
| | - Adriana T. Moreno
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil, 1500, 05509-900, São Paulo, SP, Brazil
| | - Daniela M. Ferreira
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil, 1500, 05509-900, São Paulo, SP, Brazil
| | - Fabiana C. Pimenta
- Instituto de Patologia Tropical e Saude Publica, Universidade Federal de Goias, Goiania, Brazil
| | | | - Alexandre P. Y. Lopes
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil, 1500, 05509-900, São Paulo, SP, Brazil
| | - Luciana C. C. Leite
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil, 1500, 05509-900, São Paulo, SP, Brazil
| | - Eliane N. Miyaji
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil, 1500, 05509-900, São Paulo, SP, Brazil
| |
Collapse
|
47
|
Ochs MM, Bartlett W, Briles DE, Hicks B, Jurkuvenas A, Lau P, Ren B, Millar A. Vaccine-induced human antibodies to PspA augment complement C3 deposition on Streptococcus pneumoniae. Microb Pathog 2007; 44:204-14. [PMID: 18006268 DOI: 10.1016/j.micpath.2007.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 09/25/2007] [Accepted: 09/27/2007] [Indexed: 01/09/2023]
Abstract
Pneumococcal surface protein (PspA) is a virulence factor expressed by all clinical isolates of Streptococcus pneumoniae. PspAs are variable in structure and have been grouped into clades and cross-reacting families based on sequence similarities and immunologic cross-reactivity. At least 98% of PspAs are found in PspA families 1 or 2. PspA has been shown to interfere with complement deposition on pneumococci, thus reducing opsonization and clearance of bacteria by the host immune system. Prior studies using pooled human sera have shown that PspA interferes with C3 deposition on a single strain of S. pneumoniae, WU2, and that mouse antibody to PspA can enhance the deposition of C3 on WU2. The present studies have demonstrated that these previous findings are representative of most normal human sera and each of seven different strains of S. pneumoniae. It was observed that PspAs of PspA families 1 and 2 could inhibit C3 deposition in the presence of immunoglobulin present in all but 3 of 22 normal human sera. These studies have also demonstrated that rabbit and human antibody to PspA can enhance the deposition of C3 on pneumococci expressing either family 1 or 2 PspAs and either capsular types 2, 3, or 11. A vaccine candidate that can elicit immunity that neutralizes or compensates for S. pneumoniae's ability to thwart host immunity would be of value.
Collapse
|
48
|
Cao J, Chen D, Xu W, Chen T, Xu S, Luo J, Zhao Q, Liu B, Wang D, Zhang X, Shan Y, Yin Y. Enhanced protection against pneumococcal infection elicited by immunization with the combination of PspA, PspC, and ClpP. Vaccine 2007; 25:4996-5005. [PMID: 17524530 DOI: 10.1016/j.vaccine.2007.04.069] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 04/07/2007] [Accepted: 04/20/2007] [Indexed: 11/20/2022]
Abstract
Immunization with a combination of several virulence-associated proteins is one of the strategies of developing effective protein-based vaccines to enhance the protection against Streptococcus pneumoniae. In this study, we evaluated the protection effects against pneumococcal infection caused by S. pneumoniae TIGR4 in BALB/c mice immunized with either single pneumococcal surface protein A (PspA), pneumococcal surface protein C (PspC), the caseinolytic protease (ClpP) or their combinations. The median survival times for mice immunized with single antigen or their combinations were significantly longer than that for mice treated with adjuvant alone. Mice treated with a combination of three antigens survived significantly longer than those that received either single or two antigens. The highest survival rate of the various groups of mice was observed with the combination of three antigens, this survival rate was significantly different from those for mice that received either single antigen or the combinations of two antigens except the mixture of ClpP and PspA. In the experiment of passive immunization with hyperimmune serums containing their specific polyclonal antibodies (anti-PspA serum, anti-PspC serum, anti-ClpP serum), the median survival times for mice immunized with hyperimmune serums containing specific polyclonal antibodies were significantly longer than that for control mice, the treatment of serum containing only one single polyclonal antibody could not provide higher survival rate than control serum. However, the survival rates for mice treated with the serums containing combined polyclonal antibodies were significantly higher than those for mice treated with either control serum or anti-PspA serum alone. Immunization with the combination of three hyperimmune serums also provided the best protection against S. pneumoniae. Compared to mice treated with serum containing single polyclonal antibody, the survival rate for mice treated with serums containing three polyclonal antibodies was significantly higher but was not different from those for mice treated with serums containing two polyclonal antibodies. Our findings provided evidence that a mixture of PspA, PspC, and ClpP or their polyclonal antibodies could enhance the protection against pneumococcal infection acting a synergetic effect.
Collapse
Affiliation(s)
- Ju Cao
- Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine in Chongqing Medical University, Chongqing 400016, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Senkovich O, Cook WJ, Mirza S, Hollingshead SK, Protasevich II, Briles DE, Chattopadhyay D. Structure of a complex of human lactoferrin N-lobe with pneumococcal surface protein a provides insight into microbial defense mechanism. J Mol Biol 2007; 370:701-13. [PMID: 17543335 PMCID: PMC5356469 DOI: 10.1016/j.jmb.2007.04.075] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2007] [Revised: 04/23/2007] [Accepted: 04/30/2007] [Indexed: 11/24/2022]
Abstract
Human lactoferrin, a component of the innate immune system, kills a wide variety of microorganisms including the Gram positive bacteria Streptococcus pneumoniae. Pneumococcal surface protein A (PspA) efficiently inhibits this bactericidal action. The crystal structure of a complex of the lactoferrin-binding domain of PspA with the N-lobe of human lactoferrin reveals direct and specific interactions between the negatively charged surface of PspA helices and the highly cationic lactoferricin moiety of lactoferrin. Binding of PspA blocks surface accessibility of this bactericidal peptide preventing it from penetrating the bacterial membrane. Results of site-directed mutagenesis, in vitro protein binding assays and isothermal titration calorimetry measurements corroborate that the specific electrostatic interactions observed in the crystal structure represent major associations between PspA and lactoferrin. The structure provides a snapshot of the protective mechanism utilized by pathogens against the host's first line of defense. PspA represents a major virulence factor and a promising vaccine candidate. Insights from the structure of the complex have implications for designing therapeutic strategies for treatment and prevention of pneumococcal diseases that remain a major public health problem worldwide.
Collapse
MESH Headings
- Animals
- Antigens, Bacterial/chemistry
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Antigens, Surface/chemistry
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Bacterial Proteins/chemistry
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Cattle
- Crystallography, X-Ray
- Humans
- Lactoferrin/chemistry
- Lactoferrin/genetics
- Lactoferrin/immunology
- Lactoferrin/metabolism
- Membrane Fusion
- Models, Molecular
- Mutation/genetics
- Nuclear Magnetic Resonance, Biomolecular
- Protein Binding
- Protein Structure, Quaternary
- Streptococcus pneumoniae/chemistry
- Streptococcus pneumoniae/genetics
- Streptococcus pneumoniae/immunology
- Streptococcus pneumoniae/metabolism
- Structural Homology, Protein
Collapse
Affiliation(s)
- Olga Senkovich
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - William J. Cook
- Department of Pathology, University of Alabama at Birmingham, AL 35294, USA
| | - Shaper Mirza
- Department of Microbiology, University of Alabama at Birmingham, AL 35294, USA
| | | | - Irina I. Protasevich
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - David E. Briles
- Department of Microbiology, University of Alabama at Birmingham, AL 35294, USA
| | - Debasish Chattopadhyay
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, AL 35294, USA
- Department of Medicine, University of Alabama at Birmingham, AL 35294, USA
- Corresponding author:
| |
Collapse
|
50
|
He X, McDaniel LS. The genetic background of Streptococcus pneumoniae affects protection in mice immunized with PspA. FEMS Microbiol Lett 2007; 269:189-95. [PMID: 17355599 DOI: 10.1111/j.1574-6968.2006.00516.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Anti-PspA antibodies are less efficient at protecting mice against certain pneumococcal strains. Immunization with PspA from EF5668 provided better protection against WU2 (a different capsular serotype and PspA family) than against EF5668. To understand the role of the pneumococcal genetic background in anti-PspA-mediated protection, we constructed a mutant of WU2 expressing pspA from EF5668. Both passive and active immunization demonstrated that the genetic background impacted the protection mediated by anti-PspA antibodies. We localized the protection-eliciting region to the first 122 amino acid residues of the N-terminus of the alpha-helical domain of PspA/EF5668.
Collapse
Affiliation(s)
- Xiangyun He
- Department of Microbiology, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | |
Collapse
|