1
|
Takahara Y, Sumitomo T, Kono M, Takemura M, Akamatsu Y, Hirose Y, Yamaguchi M, Nakata M, Hotomi M, Kawabata S. Pneumolysin contributes to dysfunction of nasal epithelial barrier for promotion of pneumococcal dissemination into brain tissue. mSphere 2024; 9:e0065524. [PMID: 39345124 PMCID: PMC11520308 DOI: 10.1128/msphere.00655-24] [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: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
Streptococcus pneumoniae is one of the major pathogens responsible for bacterial meningitis and neurological sequelae. The present study was conducted to identify a non-hematogenous route used by S. pneumoniae to gain access to brain tissue without causing bacteremia or pneumonia, as well as bacterial and host factors involved in this process. To investigate the molecular mechanisms and dissemination pathways of pneumococcal infection in brain tissue, mice were intranasally inoculated with S. pneumoniae strain EF3030, a clinical isolate from a patient with otitis media. Pneumococci were isolated from the frontal olfactory bulb, caudal cerebrum, and cerebellum, with neither bacteremia nor pneumonia observed in the present model. Immunostaining imaging revealed the presence of S. pneumoniae organisms in olfactory nerve fibers. Knockout of the ply gene encoding pneumolysin (PLY) markedly compromised the ability of the bacterial organisms to disseminate into brain tissue, whereas the dissemination efficiency of the complemented strain was restored to nearly the same level as the wild type. Notably, distinct upregulation of Gli1 and Snail1, which are involved in the transcriptional repression of junctional proteins, along with downregulation of E-cadherin, was detected in nasal lavage samples from mice infected with the wild-type or complemented strain, but not in those from mice infected with the ply mutant. Taken together, the present findings indicate that PLY induces Gli1-Snail1-dependent dysfunction of the nasal epithelial barrier, thus allowing pneumococcal dissemination to brain tissue that occurs in a non-hematogenous manner.IMPORTANCEBacterial meningitis, considered to be caused by bacteremia, can lead to blood-brain barrier disruption and bacterial dissemination into the central nervous system. Despite the availability of intravenously administered antibiotics with cerebrospinal fluid transferability, bacterial meningitis remains associated with high rates of morbidity and mortality. Here, we utilized Streptococcus pneumoniae strain EF3030, clinically isolated from otitis media, for the construction of a murine infection model to investigate the molecular mechanisms by which nasally colonized pneumococci disseminate into brain tissue. The obtained findings indicate that pneumolysin (PLY) induces Gli1-Snail1-dependent dysfunction of the nasal epithelial barrier, which facilitates pneumococcal dissemination to brain tissue in a non-hematogenous manner. Our results support the existence of an alternative route by which S. pneumoniae can reach the central nervous system and indicate the need for the development of novel therapeutic strategies, which would be an important contribution to the clinical management of bacterial meningitis.
Collapse
Affiliation(s)
- Yuki Takahara
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Fixed Prosthodontics and Orofacial Function, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Tomoko Sumitomo
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology—Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Moe Takemura
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Oral Surgery, Rinku General Medical Center, Izumisano, Osaka, Japan
| | - Yukako Akamatsu
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Division of Special Care Dentistry, Osaka University Dental Hospital, Osaka, Japan
| | - Yujiro Hirose
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Bioinformatics Research Unit, Osaka University Graduate School of Dentistry, Osaka, Japan
- Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Osaka, Japan
| | - Masanobu Nakata
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology—Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Osaka, Japan
| |
Collapse
|
2
|
Wang Y, Shi G, Wang X, Xie Z, Gou J, Huang L, Huang H, You W, Wang R, Yang Y, Wang F, Zhu T, Zhao D. Preliminary Evaluation of the Safety and Immunogenicity of a Novel Protein-Based Pneumococcal Vaccine in Healthy Adults Aged 18-49: A Phase Ia Randomized, Double Blind, Placebo-Controlled Clinical Study. Vaccines (Basel) 2024; 12:827. [PMID: 39203953 PMCID: PMC11358999 DOI: 10.3390/vaccines12080827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 09/03/2024] Open
Abstract
Background: Protein-based pneumococcal vaccines (PBPVs) may offer expanded protection against Streptococcus pneumoniae and tackle the antimicrobial resistance crisis in pneumococcal infections. This study examined the safety and immunogenicity in healthy adults vaccinated with three doses of a protein-based pneumococcal vaccine containing pneumococcal surface protein A (PspA) (PRX1, P3296 and P5668) and in combination with a recombinant detoxified pneumolysin protein (PlyLD). Methods: This phase Ia randomized, double blind, placebo-controlled clinical study enrolled healthy adults aged 18-49 years. The participants were randomized into experimental (low-dose, medium-dose, high-dose) and placebo groups in a ratio of 3:1. Three doses of investigational vaccine were given to the participants with an interval of two months. Safety endpoints included the occurrence of total adverse reactions, solicited local and systemic adverse reactions, unsolicited adverse reactions, serious adverse events (SAEs), and several laboratory parameters. Immunogenicity endpoints included geometric mean titers (GMT) of anti-PspA (PRX1, P3296 and P5668) and anti-PlyLD antibodies level as determined by ELISA, seropositivity rates of PspA and PlyLD antibodies (>4-fold increase) and neutralization activity of anti-Ply antibody in serum. Results: A total of 118 participants completed the study of three doses. The candidate PBPV was safe and well-tolerated in all experimental groups. No vaccine-related SAEs were observed in this study. Most solicited adverse reactions were mild and transient. The most frequently reported solicited adverse reactions in the medium- and high-dose groups was pain at the injection site, while in the low-dose group it was elevated blood pressure. The immunogenicity data showed a sharp increase in the GMT level of anti-PspA-RX1, anti-PspA-3296, anti-PspA-5668, and anti-PlyLD antibodies in serum. The results also showed that the elicited antibodies were dosage-dependent. The high-dose group showed a higher immune response against PspA-RX1, PspA-3296, PspA-5668, and PlyLD antigens. However, repeat vaccination did not increase the level of anti-PspA antibodies but the level of anti-PlyLD antibody. High seropositivity rates were also observed for anti-PspA-RX1, anti-PspA-3296, anti-PspA-5668, and anti-PlyLD antibodies. In addition, a significant difference in the GMT levels of anti-Ply antibody between the high-, medium-, and low-dose groups post each vaccination were indicated by neutralization activity tests. Conclusions: The PBPV showed a safe and immunogenic profile in this clinical trial. Taking into consideration both safety and immunogenicity data, we propose a single dose of 50 µg (medium dose) of PBPV as the optimum approach in providing expanded protection against Streptococcus pneumoniae.
Collapse
Affiliation(s)
- Yanxia Wang
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Gang Shi
- National Institutes for Food and Drug Control, Beijing 100050, China;
| | - Xue Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Zhiqiang Xie
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Jinbo Gou
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Lili Huang
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Haitao Huang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Wangyang You
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Ruijie Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Yongli Yang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China;
| | - Feiyu Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Tao Zhu
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Dongyang Zhao
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| |
Collapse
|
3
|
Gupta LK, Molla J, Prabhu AA. Story of Pore-Forming Proteins from Deadly Disease-Causing Agents to Modern Applications with Evolutionary Significance. Mol Biotechnol 2024; 66:1327-1356. [PMID: 37294530 DOI: 10.1007/s12033-023-00776-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/21/2023] [Indexed: 06/10/2023]
Abstract
Animal venoms are a complex mixture of highly specialized toxic molecules. Among them, pore-forming proteins (PFPs) or toxins (PFTs) are one of the major disease-causing toxic elements. The ability of the PFPs in defense and toxicity through pore formation on the host cell surface makes them unique among the toxin proteins. These features made them attractive for academic and research purposes for years in the areas of microbiology as well as structural biology. All the PFPs share a common mechanism of action for the attack of host cells and pore formation in which the selected pore-forming motifs of the host cell membrane-bound protein molecules drive to the lipid bilayer of the cell membrane and eventually produces water-filled pores. But surprisingly their sequence similarity is very poor. Their existence can be seen both in a soluble state and also in transmembrane complexes in the cell membrane. PFPs are prevalent toxic factors that are predominately produced by all kingdoms of life such as virulence bacteria, nematodes, fungi, protozoan parasites, frogs, plants, and also from higher organisms. Nowadays, multiple approaches to applications of PFPs have been conducted by researchers both in basic as well as applied biological research. Although PFPs are very devastating for human health nowadays researchers have been successful in making these toxic proteins into therapeutics through the preparation of immunotoxins. We have discussed the structural, and functional mechanism of action, evolutionary significance through dendrogram, domain organization, and practical applications for various approaches. This review aims to emphasize the PFTs to summarize toxic proteins together for basic knowledge as well as to highlight the current challenges, and literature gap along with the perspective of promising biotechnological applications for their future research.
Collapse
Affiliation(s)
- Laxmi Kumari Gupta
- Bioprocess Development Laboratory, Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India
| | - Johiruddin Molla
- Ghatal Rabindra Satabarsiki Mahavidyalaya Ghatal, Paschim Medinipur, Ghatal, West Bengal, 721212, India
| | - Ashish A Prabhu
- Bioprocess Development Laboratory, Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India.
| |
Collapse
|
4
|
Zahid A, Wilson JC, Grice ID, Peak IR. Otitis media: recent advances in otitis media vaccine development and model systems. Front Microbiol 2024; 15:1345027. [PMID: 38328427 PMCID: PMC10847372 DOI: 10.3389/fmicb.2024.1345027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Otitis media is an inflammatory disorder of the middle ear caused by airways-associated bacterial or viral infections. It is one of the most common childhood infections as globally more than 80% of children are diagnosed with acute otitis media by 3 years of age and it is a common reason for doctor's visits, antibiotics prescriptions, and surgery among children. Otitis media is a multifactorial disease with various genetic, immunologic, infectious, and environmental factors predisposing children to develop ear infections. Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis are the most common culprits responsible for acute otitis media. Despite the massive global disease burden, the pathogenesis of otitis media is still unclear and requires extensive future research. Antibiotics are the preferred treatment to cure middle ear infections, however, the antimicrobial resistance rate of common middle ear pathogens has increased considerably over the years. At present, pneumococcal and influenza vaccines are administered as a preventive measure against otitis media, nevertheless, these vaccines are only beneficial in preventing carriage and/or disease caused by vaccine serotypes. Otitis media caused by non-vaccine serotype pneumococci, non-typeable H. influenza, and M. catarrhalis remain an important healthcare burden. The development of multi-species vaccines is an arduous process but is required to reduce the global burden of this disease. Many novel vaccines against S. pneumoniae, non-typeable H. influenza, and M. catarrhalis are in preclinical trials. It is anticipated that these vaccines will lower the disease burden and provide better protection against otitis media. To study disease pathology the rat, mouse, and chinchilla are commonly used to induce experimental acute otitis media to test new therapeutics, including antibiotics and vaccines. Each of these models has its advantages and disadvantages, yet there is still a need to develop an improved animal model providing a better correlated mechanistic understanding of human middle ear infections, thereby underpinning the development of more effective otitis media therapeutics. This review provides an updated summary of current vaccines against otitis media, various animal models of otitis media, their limitations, and some future insights in this field providing a springboard in the development of new animal models and novel vaccines for otitis media.
Collapse
Affiliation(s)
- Ayesha Zahid
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Jennifer C. Wilson
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD, Australia
| | - I. Darren Grice
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD, Australia
| | - Ian R. Peak
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
5
|
Duke JA, Avci FY. Emerging vaccine strategies against the incessant pneumococcal disease. NPJ Vaccines 2023; 8:122. [PMID: 37591986 PMCID: PMC10435554 DOI: 10.1038/s41541-023-00715-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The incidence of invasive pneumococcal disease (IPD) caused by infection with the pathogen Streptococcus pneumoniae (Spn) has been on a downward trend for decades due to worldwide vaccination programs. Despite the clinical successes observed, the Center for Disease Control (CDC) reports that the continued global burden of S. pneumoniae will be in the millions each year, with a case-fatality rate hovering around 5%. Thus, it is a top priority to continue developing new Spn vaccination strategies to harness immunological insight and increase the magnitude of protection provided. As emphasized by the World Health Organization (WHO), it is also crucial to broaden the implementation of vaccines that are already obtainable in the clinical setting. This review focuses on the immune mechanisms triggered by existing pneumococcal vaccines and provides an overview of the current and upcoming clinical strategies being employed. We highlight the associated challenges of serotype selectivity and using pneumococcal-derived proteins as alternative vaccine antigens.
Collapse
Affiliation(s)
- Jeremy A Duke
- Sanofi, Suite 300, 2501 Discovery Drive, Orlando, FL, 32826, USA
| | - Fikri Y Avci
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
6
|
Silva PH, Vázquez Y, Campusano C, Retamal-Díaz A, Lay MK, Muñoz CA, González PA, Kalergis AM, Bueno SM. Non-capsular based immunization approaches to prevent Streptococcus pneumoniae infection. Front Cell Infect Microbiol 2022; 12:949469. [PMID: 36225231 PMCID: PMC9548657 DOI: 10.3389/fcimb.2022.949469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive bacterium and the leading cause of bacterial pneumonia in children and the elderly worldwide. Currently, two types of licensed vaccines are available to prevent the disease caused by this pathogen: the 23-valent pneumococcal polysaccharide-based vaccine and the 7-, 10, 13, 15 and 20-valent pneumococcal conjugate vaccine. However, these vaccines, composed of the principal capsular polysaccharide of leading serotypes of this bacterium, have some problems, such as high production costs and serotype-dependent effectiveness. These drawbacks have stimulated research initiatives into non-capsular-based vaccines in search of a universal vaccine against S. pneumoniae. In the last decades, several research groups have been developing various new vaccines against this bacterium based on recombinant proteins, live attenuated bacterium, inactivated whole-cell vaccines, and other newer platforms. Here, we review and discuss the status of non-capsular vaccines against S. pneumoniae and the future of these alternatives in a post-pandemic scenario.
Collapse
Affiliation(s)
- Pedro H. Silva
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Campusano
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Margarita K. Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Christian A. Muñoz
- Unidad de Microbiología, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Susan M. Bueno,
| |
Collapse
|
7
|
Aceil J, Avci FY. Pneumococcal Surface Proteins as Virulence Factors, Immunogens, and Conserved Vaccine Targets. Front Cell Infect Microbiol 2022; 12:832254. [PMID: 35646747 PMCID: PMC9133333 DOI: 10.3389/fcimb.2022.832254] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen that causes over 1 million deaths annually despite the availability of several multivalent pneumococcal conjugate vaccines (PCVs). Due to the limitations surrounding PCVs along with an evolutionary rise in antibiotic-resistant and unencapsulated strains, conserved immunogenic proteins as vaccine targets continue to be an important field of study for pneumococcal disease prevention. In this review, we provide an overview of multiple classes of conserved surface proteins that have been studied for their contribution to pneumococcal virulence. Furthermore, we discuss the immune responses observed in response to these proteins and their promise as vaccine targets.
Collapse
|
8
|
Pereira JM, Xu S, Leong JM, Sousa S. The Yin and Yang of Pneumolysin During Pneumococcal Infection. Front Immunol 2022; 13:878244. [PMID: 35529870 PMCID: PMC9074694 DOI: 10.3389/fimmu.2022.878244] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Pneumolysin (PLY) is a pore-forming toxin produced by the human pathobiont Streptococcus pneumoniae, the major cause of pneumonia worldwide. PLY, a key pneumococcal virulence factor, can form transmembrane pores in host cells, disrupting plasma membrane integrity and deregulating cellular homeostasis. At lytic concentrations, PLY causes cell death. At sub-lytic concentrations, PLY triggers host cell survival pathways that cooperate to reseal the damaged plasma membrane and restore cell homeostasis. While PLY is generally considered a pivotal factor promoting S. pneumoniae colonization and survival, it is also a powerful trigger of the innate and adaptive host immune response against bacterial infection. The dichotomy of PLY as both a key bacterial virulence factor and a trigger for host immune modulation allows the toxin to display both "Yin" and "Yang" properties during infection, promoting disease by membrane perforation and activating inflammatory pathways, while also mitigating damage by triggering host cell repair and initiating anti-inflammatory responses. Due to its cytolytic activity and diverse immunomodulatory properties, PLY is integral to every stage of S. pneumoniae pathogenesis and may tip the balance towards either the pathogen or the host depending on the context of infection.
Collapse
Affiliation(s)
- Joana M. Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Molecular and Cellular (MC) Biology PhD Program, ICBAS - Instituto de Ciência Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, United States
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Parveen S, Subramanian K. Emerging Roles of Extracellular Vesicles in Pneumococcal Infections: Immunomodulators to Potential Novel Vaccine Candidates. Front Cell Infect Microbiol 2022; 12:836070. [PMID: 35237534 PMCID: PMC8882830 DOI: 10.3389/fcimb.2022.836070] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
The Gram-positive bacterial pathogen, Streptococcus pneumoniae is a major global health threat that kills over one million people worldwide. The pneumococcus commonly colonizes the nasopharynx asymptomatically as a commensal, but is also capable of causing a wide range of life-threatening diseases such as pneumonia, meningitis and septicemia upon migration into the lower respiratory tract and spread to internal organs. Emergence of antibiotic resistant strains and non-vaccine serotypes has led to the classification of pneumococcal bacteria as a priority pathogen by the World Health Organization that needs urgent research into bacterial pathogenesis and development of novel vaccine strategies. Extracellular vesicles are spherical membrane bound structures that are released by both pathogen and host cells, and influence bacterial pathogenesis as well as the immune response. Recent studies have found that while bacterial vesicles shuttle virulence factors and toxins into host cells and regulate inflammatory responses, vesicles released from the infected host cells contain both bacterial and host proteins that are antigenic and immunomodulatory. Bacterial membrane vesicles have great potential to be developed as cell-free vaccine candidates in the future due to their immunogenicity and biostability. Host-derived vesicles isolated from patient biofluids such as blood and bronchoalveolar lavage could be used to identify potential diagnostic biomarkers as well as engineered to deliver desired payloads to specific target cells for immunotherapy. In this review, we summarize the recent developments on the role of bacterial and host vesicles in pneumococcal infections and future prospects in developing novel therapeutics and diagnostics for control of invasive pneumococcal diseases.
Collapse
|
10
|
Phelps CC, Vadia S, Boyaka PN, Varikuti S, Attia Z, Dubey P, Satoskar AR, Tweten R, Seveau S. A listeriolysin O subunit vaccine is protective against Listeria monocytogenes. Vaccine 2020; 38:5803-5813. [PMID: 32684498 DOI: 10.1016/j.vaccine.2020.06.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 05/12/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Listeria monocytogenes is a facultative intracellular pathogen responsible for the life-threatening disease listeriosis. The pore-forming toxin listeriolysin O (LLO) is a critical virulence factor that plays a major role in the L. monocytogenes intracellular lifecycle and is indispensable for pathogenesis. LLO is also a dominant antigen for T cells involved in sterilizing immunity and it was proposed that LLO acts as a T cell adjuvant. In this work, we generated a novel full-length LLO toxoid (LLOT) in which the cholesterol-recognition motif, a threonine-leucine pair located at the tip of the LLO C-terminal domain, was substituted with two glycine residues. We showed that LLOT lost its ability to bind cholesterol and to form pores. Importantly, LLOT retained binding to the surface of epithelial cells and macrophages, suggesting that it could efficiently be captured by antigen-presenting cells. We then determined if LLOT can be used as an antigen and adjuvant to protect mice from L. monocytogenes infection. Mice were immunized with LLOT alone or together with cholera toxin or Alum as adjuvants. We found that mice immunized with LLOT alone or in combination with the Th2-inducing adjuvant Alum were not protected against L. monocytogenes. On the other hand, mice immunized with LLOT along with the experimental adjuvant cholera toxin, were protected against L. monocytogenes, as evidenced by a significant decrease in bacterial burden in the liver and spleen three days post-infection. This immunization regimen elicited mixed Th1, Th2, and Th17 responses, as well as the generation of LLO-neutralizing antibodies. Further, we identified T cells as being required for immunization-induced reductions in bacterial burden, whereas B cells were dispensable in our model of non-pregnant young mice. Overall, this work establishes that LLOT is a promising vaccine antigen for the induction of protective immunity against L. monocytogenes by subunit vaccines containing Th1-driving adjuvants.
Collapse
Affiliation(s)
- Christopher C Phelps
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA
| | - Stephen Vadia
- Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA; Department of Biology, Washington University in St. Louis, MO 63130, USA
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Rodney Tweten
- Department of Microbial & Immunology, University of Oklahoma, Oklahoma City, OK, USA
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
11
|
Nishimoto AT, Rosch JW, Tuomanen EI. Pneumolysin: Pathogenesis and Therapeutic Target. Front Microbiol 2020; 11:1543. [PMID: 32714314 PMCID: PMC7343714 DOI: 10.3389/fmicb.2020.01543] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen responsible for widespread illness and is a major global health issue for children, the elderly, and the immunocompromised population. Pneumolysin (PLY) is a cholesterol-dependent cytolysin (CDC) and key pneumococcal virulence factor involved in all phases of pneumococcal disease, including transmission, colonization, and infection. In this review we cover the biology and cytolytic function of PLY, its contribution to S. pneumoniae pathogenesis, and its known interactions and effects on the host with regard to tissue damage and immune response. Additionally, we review statins as a therapeutic option for CDC toxicity and PLY toxoid as a vaccine candidate in protein-based vaccines.
Collapse
Affiliation(s)
- Andrew T Nishimoto
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Jason W Rosch
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Elaine I Tuomanen
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
12
|
Thanawastien A, Joyce KE, Cartee RT, Haines LA, Pelton SI, Tweten RK, Killeen KP. Preclinical in vitro and in vivo profile of a highly-attenuated, broadly efficacious pneumolysin genetic toxoid. Vaccine 2020; 39:1652-1660. [PMID: 32532546 PMCID: PMC8237519 DOI: 10.1016/j.vaccine.2020.04.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/03/2020] [Accepted: 04/26/2020] [Indexed: 02/05/2023]
Abstract
Pneumolysin is a highly conserved, cholesterol-dependent cytolysin that is an important Streptococcus pneumoniae virulence factor and an attractive target for vaccine development. To attenuate pneumolysin toxicity, a genetic toxoid was constructed with two amino acid changes, G293S and L460D, termed PLY-D, that reduced cytolytic activity > 125,000-fold. In mice, PLY-D elicited high anti-PLY IgG antibody titers that neutralized the cytolytic activity of the wild-type toxin in vitro. To evaluate the protective efficacy of PLY-D, mice were immunized intramuscularly and then challenged intranasally with a lethal dose of 28 clinical isolates of S. pneumoniae originating from different geographical locations, disease states (i.e. bacteremia, pneumonia), or body sites (i.e. sputum, blood). PLY-D immunization conferred significant protection from challenge with 17 of 20 serotypes (85%) and 22 of 28 strains (79%). Further, we demonstrated that immunization with PLY-D provided statistically significant improvement in survival against challenge with serotype 4 and 18C strains compared to mice immunized with a pneumococcal conjugate vaccine Prevnar 13® (PCV13). Co-administration of PLY-D and PCV13 conferred greater protection against challenge with a serotype 6B strain than immunization with either vaccine alone. These data indicate that PLY-D is a broadly protective antigen with the potential to serve as a serotype-independent vaccine against invasive pneumococcal disease either alone or in combination with PCVs.
Collapse
Affiliation(s)
- Ann Thanawastien
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Kelsey E Joyce
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Robert T Cartee
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Laurel A Haines
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Stephen I Pelton
- Boston University Schools of Medicine and Public Health, Boston Medical Center, Boston, MA, United States
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Kevin P Killeen
- Matrivax Research & Development Corporation, Boston, MA, United States.
| |
Collapse
|
13
|
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: 9.2] [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
|
14
|
Hammitt LL, Campbell JC, Borys D, Weatherholtz RC, Reid R, Goklish N, Moulton LH, Traskine M, Song Y, Swinnen K, Santosham M, O'Brien KL. Efficacy, safety and immunogenicity of a pneumococcal protein-based vaccine co-administered with 13-valent pneumococcal conjugate vaccine against acute otitis media in young children: A phase IIb randomized study. Vaccine 2019; 37:7482-7492. [PMID: 31629570 DOI: 10.1016/j.vaccine.2019.09.076] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/18/2019] [Accepted: 09/20/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Native American populations experience a substantial burden of pneumococcal disease despite use of highly effective pneumococcal conjugate vaccines (PCVs). Protein-based pneumococcal vaccines may extend protection beyond the serotype-specific protection elicited by PCVs. METHODS In this phase IIb, double-blind, controlled trial, 6-12 weeks-old Native American infants randomized 1:1, received either a protein-based pneumococcal vaccine (dPly/PhtD) containing pneumolysin toxoid (dPly, 10 µg) and pneumococcal histidine triad protein D (PhtD, 10 µg) or placebo, administered along with 13-valent PCV (PCV13) at ages 2, 4, 6 and 12-15 months. Other pediatric vaccines were given per the routine immunization schedule. We assessed vaccine efficacy (VE) against acute otitis media (AOM) and acute lower respiratory tract infection (ALRI) endpoints. Immunogenicity, reactogenicity and unsolicited adverse events were assessed in a sub-cohort and serious adverse events were assessed in all children. RESULTS 1803 infants were randomized (900 dPly/PhtD; 903 Control). VE against all episodes of American Academy of Pediatrics (AAP)-defined AOM was 3.8% (95% confidence interval: -11.4, 16.9). Point estimates of VE against other AOM outcomes ranged between 2.9% (-9.5, 14.0) and 5.2% (-8.0, 16.8). Point estimates of VE against ALRI outcomes ranged between -4.4% (-39.2, 21.8) and 2.0% (-18.3, 18.8). Point estimates of VE tended to be higher against first than all episodes but the confidence intervals included zero. dPly/PhtD vaccine was immunogenic and had an acceptable reactogenicity and safety profile after primary and booster vaccination in Native American infants. CONCLUSIONS The dPly/PhtD vaccine was immunogenic and well tolerated, however, incremental efficacy in preventing AAP-AOM over PCV13 was not demonstrated. CLINICAL TRIALS REGISTRATION NCT01545375 (www.clinicaltrials.gov).
Collapse
Affiliation(s)
- Laura L Hammitt
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.
| | - James C Campbell
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Robert C Weatherholtz
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Raymond Reid
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Novalene Goklish
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lawrence H Moulton
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Yue Song
- XPE Pharma & Science c/o GSK, Wavre, Belgium
| | | | - Mathuram Santosham
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Katherine L O'Brien
- Center for American Indian Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
15
|
Pharmacological Targeting of Pore-Forming Toxins as Adjunctive Therapy for Invasive Bacterial Infection. Toxins (Basel) 2018; 10:toxins10120542. [PMID: 30562923 PMCID: PMC6316385 DOI: 10.3390/toxins10120542] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/23/2022] Open
Abstract
For many of the most important human bacterial infections, invasive disease severity is fueled by the cell damaging and pro-inflammatory effects of secreted pore-forming toxins (PFTs). Isogenic PFT-knockout mutants, e.g., Staphylococcus aureus lacking α-toxin or Streptococcus pneumoniae deficient in pneumolysin, show attenuation in animal infection models. This knowledge has inspired multi-model investigations of strategies to neutralize PFTs or counteract their toxicity as a novel pharmacological approach to ameliorate disease pathogenesis in clinical disease. Promising examples of small molecule, antibody or nanotherapeutic drug candidates that directly bind and neutralize PFTs, block their oligomerization or membrane receptor interactions, plug establishment membrane pores, or boost host cell resiliency to withstand PFT action have emerged. The present review highlights these new concepts, with a special focus on β-PFTs produced by leading invasive human Gram-positive bacterial pathogens. Such anti-virulence therapies could be applied as an adjunctive therapy to antibiotic-sensitive and -resistant strains alike, and further could be free of deleterious effects that deplete the normal microflora.
Collapse
|
16
|
Nagai K, Domon H, Maekawa T, Hiyoshi T, Tamura H, Yonezawa D, Habuka R, Saitoh A, Terao Y. Immunization with pneumococcal elongation factor Tu enhances serotype-independent protection against Streptococcus pneumoniae infection. Vaccine 2018; 37:160-168. [PMID: 30442480 DOI: 10.1016/j.vaccine.2018.11.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/01/2018] [Accepted: 11/07/2018] [Indexed: 11/25/2022]
Abstract
Vaccination is an effective strategy to prevent pneumococcal diseases. Currently, licensed vaccines include the pneumococcal polysaccharide vaccine (PPSV) and the pneumococcal conjugate vaccine (PCV), which target some of the most common of the 94 serotypes of S. pneumoniae based on their capsular composition. However, it has been reported that PPSV is not effective in children aged less than 2 years old and PCV induces serotype replacement, which means that the pneumococcal population has changed following widespread introduction of these vaccines, and the non-vaccine serotypes have increased in being the cause of invasive pneumococcal disease. Therefore, it is important that there is development of novel pneumococcal vaccines to either replace or complement current polysaccharide-based vaccines. Our previous study suggested that S. pneumoniae releases elongation factor Tu (EF-Tu) through autolysis followed by the induction of proinflammatory cytokines in macrophages via toll-like receptor 4, that may contribute to the development of pneumococcal diseases. In this study, we investigated the expression of EF-Tu in various S. pneumoniae strains and whether EF-Tu could be an antigen candidate for serotype-independent vaccine against pneumococcal infection. Western blotting and flow cytometry analysis revealed that EF-Tu is a common factor expressed on the surface of all pneumococcal strains tested, as well as intracellularly. In addition, we demonstrate that immunization with recombinant (r) EF-Tu induced the production of inflammatory cytokines and the IgG1 and IgG2a antibodies in mice, and increased the CD4+ T-cells proportion in splenocytes. We also reveal that anti-EF-Tu serum increased the phagocytic activity of mouse peritoneal macrophages against S. pneumoniae infection, independent of their serotypes. Finally, our results indicate that mice immunized with rEF-Tu were significantly and non-specifically protected against lethal challenges with S. pneumoniae serotypes (2 and 15A). Therefore, pneumococcal EF-Tu could be an antigen candidate for the serotype-independent vaccine against pneumococcal infection.
Collapse
Affiliation(s)
- Kosuke Nagai
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Research Centre for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoki Maekawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Research Centre for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takumi Hiyoshi
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hikaru Tamura
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Research Centre for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Daisuke Yonezawa
- Research Centre for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Rie Habuka
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Akihiko Saitoh
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Research Centre for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
17
|
Immunogenicity and mechanisms of action of PnuBioVax, a multi-antigen serotype-independent prophylactic vaccine against infection with Streptococcus pneumoniae. Vaccine 2018; 36:4255-4264. [PMID: 29895498 DOI: 10.1016/j.vaccine.2018.05.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 11/21/2022]
Abstract
Streptococcus pneumoniae has multiple protein antigens on the surface in addition to the serotype specific polysaccharide capsule antigen. Whilst the capsule antigen is the target of the polysaccharide vaccines, bacterial proteins can also act as targets for the immune system. PnuBioVax (PBV) is being developed as a multi-antigen, serotype-independent prophylactic vaccine against S. pneumoniae disease. In this study we have sought to elucidate the immune response to PBV in immunised rabbits. Sera from PBV immunised rabbits contained high levels of IgG antibodies to the PBV vaccine, and pneumococcal antigens PspA, Ply, PsaA and PiuA which are components of PBV, when compared with control sera. The PBV sera supported killing of the vaccine strain TIGR4 in an opsonophagocytic killing assay and heterologous strains 6B, 19F and 15B. In addition, incubation in PBV sera led to agglutination of several strains of pneumococci, inhibition of Ply-mediated lysis of erythrocytes and reduced bacterial invasion of lung epithelial cells in vitro. These data suggest that PBV vaccination generates sera that has multiple mechanisms of action that may provide effective protection against pneumococcal infection and give broader strain coverage than the current polysaccharide based vaccines.
Collapse
|
18
|
Ritchie ND, Ritchie R, Bayes HK, Mitchell TJ, Evans TJ. IL-17 can be protective or deleterious in murine pneumococcal pneumonia. PLoS Pathog 2018; 14:e1007099. [PMID: 29813133 PMCID: PMC5993294 DOI: 10.1371/journal.ppat.1007099] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/08/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Streptococcus pneumoniae is the major bacterial cause of community-acquired pneumonia, and the leading agent of childhood pneumonia deaths worldwide. Nasal colonization is an essential step prior to infection. The cytokine IL-17 protects against such colonization and vaccines that enhance IL-17 responses to pneumococcal colonization are being developed. The role of IL-17 in host defence against pneumonia is not known. To address this issue, we have utilized a murine model of pneumococcal pneumonia in which the gene for the IL-17 cytokine family receptor, Il17ra, has been inactivated. Using this model, we show that IL-17 produced predominantly from γδ T cells protects mice against death from the invasive TIGR4 strain (serotype 4) which expresses a relatively thin capsule. However, in pneumonia produced by two heavily encapsulated strains with low invasive potential (serotypes 3 and 6B), IL-17 significantly enhanced mortality. Neutrophil uptake and killing of the serotype 3 strain was significantly impaired compared to the serotype 4 strain and depletion of neutrophils with antibody enhanced survival of mice infected with the highly encapsulated SRL1 strain. These data strongly suggest that IL-17 mediated neutrophil recruitment to the lungs clears infection from the invasive TIGR4 strain but that lung neutrophils exacerbate disease caused by the highly encapsulated pneumococcal strains. Thus, whilst augmenting IL-17 immune responses against pneumococci may decrease nasal colonization, this may worsen outcome during pneumonia caused by some strains.
Collapse
MESH Headings
- Animals
- Bacteremia/immunology
- Bacteremia/microbiology
- Bacterial Capsules/immunology
- Bacterial Capsules/ultrastructure
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoalveolar Lavage Fluid/microbiology
- Disease Models, Animal
- Interleukin-17/immunology
- Lung/cytology
- Lung/enzymology
- Lung/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron, Transmission
- Microscopy, Fluorescence
- Nasopharynx/microbiology
- Neutrophils/cytology
- Neutrophils/immunology
- Peroxidase/metabolism
- Phagocytosis
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/mortality
- Pneumonia, Pneumococcal/prevention & control
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Interleukin-17/genetics
- Specific Pathogen-Free Organisms
- Streptococcus pneumoniae/immunology
- Streptococcus pneumoniae/ultrastructure
Collapse
Affiliation(s)
- Neil D. Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Ryan Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Hannah K. Bayes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Tim J. Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences University of Birmingham, Birmingham, United Kingdom
| | - Tom J. Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
19
|
Immunodominance in T cell responses elicited against different domains of detoxified pneumolysin PlyD1. PLoS One 2018; 13:e0193650. [PMID: 29509778 PMCID: PMC5839544 DOI: 10.1371/journal.pone.0193650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/15/2018] [Indexed: 12/05/2022] Open
Abstract
Detoxified pneumolysin, PlyD1, is a protein vaccine candidate that induces protection against infections with Streptococcus pneumoniae in mouse models. Despite extensive knowledge on antibody responses against PlyD1, limited information is available about PlyD1 induced T cell recognition. Here we interrogated epitope breadth and functional characteristics of the T cell response to PlyD1 in two mouse strains. BALB/c (H-2d) and C57BL/6 (H-2b) mice were vaccinated with Al(OH)3-adjuvanted or non-adjuvanted PlyD1, or placebo, on day 0, 21 and 42 and were sacrificed at day 56 for collection of sera and spleens. Vaccination with adjuvanted and non-adjuvanted PlyD1 induced anti-pneumolysin IgG antibodies with neutralizing capacity in both mouse strains. Adjuvantation of PlyD1 enhanced the serological responses in both strains. In vitro restimulation of splenocytes with PlyD1 and 18-mer synthetic peptides derived from pneumolysin revealed specific proliferative and cytokine responses. For both mouse strains, one immunodominant and three subdominant natural epitopes were identified. Overlap between H-2d and H-2b restricted T cell epitopes was limited, yet similarities were found between epitopes processed in mice and predicted to be immunogenic in humans. H-2d restricted T cell epitopes were localized in pneumolysin domains 2 and 3, whereas H-2b epitopes were scattered over the protein. Cytokine responses show mostly a Th2 profile, with low levels of Th1 cytokines, in both mouse strains. In conclusion, PlyD1 evokes T cell responses in mice directed against multiple epitope regions, that is dependent on Major Histocompatibility Complex (MHC) background. These results are important to understand human PlyD1 T cell immunogenicity, to guide cell mediated immunity studies in the context of vaccine development.
Collapse
|
20
|
Entwisle C, Hill S, Pang Y, Joachim M, McIlgorm A, Colaco C, Goldblatt D, De Gorguette D'Argoeuves P, Bailey C. Safety and immunogenicity of a novel multiple antigen pneumococcal vaccine in adults: A Phase 1 randomised clinical trial. Vaccine 2017; 35:7181-7186. [PMID: 29132988 DOI: 10.1016/j.vaccine.2017.10.076] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pneumococcal vaccines, combining multiple protein antigens, provide an alternative approach to currently marketed vaccines and may provide broader protection against pneumococcal disease. This trial evaluated the safety and immunogenicity of a novel vaccine candidate PnuBioVax in healthy young adults. METHODS In a Phase 1 double-blind study, 36 subjects (18-40 years) were randomised to receive 3 doses of PnuBioVax, 28 days apart, at one of three dose levels (50, 200, 500 µg) or placebo. Safety assessments included rates of emergent adverse events (AEs), injection site and systemic reactions. Immunogenicity endpoints included antibody titre against PnuBioVax and selected pneumococcal antigens. RESULTS In the placebo (n=9) and PnuBioVax (n=27) vaccinated subjects, there were 15 and 72, reported TEAEs, respectively. The majority of TEAEs were classified as common vaccine related AEs. There were no serious AEs. Common vaccine-related AEs occurred in 13 PnuBioVax (48%) and 2 placebo (22%) subjects and were all headaches (mild and moderate). Injection site reactions, mostly pain and tenderness (graded mild or moderate) were reported, in particular in the 200 µg and 500 µg PnuBioVax groups. There were no clinically significant changes in vital signs, ECG or blood chemistries. Subjects receiving the higher dose (200 and 500 μg) demonstrated a greater fold increase in IgG titre compared with the starting dose (50 μg) or the placebo group. The fold-increase was statistically significantly higher for 200 and 500µg PnuBioVax vs 50µg PnuBioVax and placebo at each timepoint post-immunisation. Most subjects receiving 200 and 500 µg PnuBioVax demonstrated a ≥2-fold increase in antibody against pneumolysin (Ply), Pneumococcal surface antigen (PsaA), PiaA (Pneumococcal iron acquisition), PspA (Pneumococcal surface protein A) and pilus proteins (RrgB and RrgA). CONCLUSIONS All dose levels were considered safe and well tolerated. There was a statistically significant increase in anti-PnuBioVax IgG titres at the 200 and 500 µg dose levels compared to 50 µg and placebo. TRIAL REGISTRATION NUMBER NCT02572635https://www.clinicaltrials.gov.
Collapse
Affiliation(s)
| | - Sue Hill
- ImmunoBiology Ltd, Babraham Research Campus, Cambridge, UK
| | - Yin Pang
- ImmunoBiology Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Ann McIlgorm
- ImmunoBiology Ltd, Babraham Research Campus, Cambridge, UK
| | - Camilo Colaco
- ImmunoBiology Ltd, Babraham Research Campus, Cambridge, UK
| | - David Goldblatt
- Immunobiology Section, UCL GOS Institute of Child Health, London, UK
| | | | - Chris Bailey
- ImmunoBiology Ltd, Babraham Research Campus, Cambridge, UK
| |
Collapse
|
21
|
Lewis ML, Surewaard BGJ. Neutrophil evasion strategies by Streptococcus pneumoniae and Staphylococcus aureus. Cell Tissue Res 2017; 371:489-503. [PMID: 29204747 DOI: 10.1007/s00441-017-2737-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/06/2017] [Indexed: 02/05/2023]
Abstract
Humans are well equipped to defend themselves against bacteria. The innate immune system employs diverse mechanisms to recognize, control and initiate a response that can destroy millions of different microbes. Microbes that evade the sophisticated innate immune system are able to escape detection and could become pathogens. The pathogens Streptococcus pneumoniae and Staphylococcus aureus are particularly successful due to the development of a wide variety of virulence strategies for bacterial pathogenesis and they invest significant efforts towards mechanisms that allow for neutrophil evasion. Neutrophils are a primary cellular defense and can rapidly kill invading microbes, which is an indispensable function for maintaining host health. This review compares the key features of Streptococcus pneumoniae and Staphylococcus aureus in epidemiology, with a specific focus on virulence mechanisms utilized to evade neutrophils in bacterial pathogenesis. It is important to understand the complex interactions between pathogenic bacteria and neutrophils so that we can disrupt the ability of pathogens to cause disease.
Collapse
Affiliation(s)
- Megan L Lewis
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Bas G J Surewaard
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada. .,Department of Medical Microbiology, University Medical Centre, Utrecht, Netherlands.
| |
Collapse
|
22
|
Principi N, Esposito S. Development of pneumococcal vaccines over the last 10 years. Expert Opin Biol Ther 2017; 18:7-17. [DOI: 10.1080/14712598.2018.1384462] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nicola Principi
- Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Susanna Esposito
- Paediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
23
|
Lu J, Hou H, Wang D, Leenhouts K, Roosmalen MLV, Sun T, Gu T, Song Y, Jiang C, Kong W, Wu Y. Systemic and mucosal immune responses elicited by intranasal immunization with a pneumococcal bacterium-like particle-based vaccine displaying pneumolysin mutant Plym2. Immunol Lett 2017; 187:41-46. [PMID: 28487097 DOI: 10.1016/j.imlet.2017.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/12/2017] [Accepted: 05/04/2017] [Indexed: 12/18/2022]
Abstract
Pneumolysin (Ply) is an important virulence factor in pneumococcal infection and a conserved cholesterol-binding cytotoxin expressed by all serotypes of Streptococcus pneumoniae. We previously developed a highly detoxified Ply mutant designated Plym2 by replacement of two amino acids (C428G and W433F), which lost cytotoxicity but retained the ability to induce neutralizing antibodies. In the present work, we applied bacterium-like particles (BLPs) as a carrier and immunostimulant for the development of a Plym2 intranasal vaccine, in which the Plym2 protein was displayed on the surface of BLPs. Intranasal immunization of mice with BLP-Plym2 not only induced a high level of serum IgG antibodies but also a high level of mucosal SIgA antibodies in lung lavages. Antiserum induced by the BLP-Plym2 vaccine elicited high-titer neutralization activity which could inhibit the hemolysis of wild-type Ply. In conclusion, the BLP-Plym2 vaccine was demonstrated to be a promising strategy for intranasal immunization to enhance both systemic and mucosal immune responses.
Collapse
Affiliation(s)
- Jingcai Lu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China; Changchun BCHT Biotechnology Co., Changchun 130012, China
| | - Hongjia Hou
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China
| | - Dandan Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China
| | - Kees Leenhouts
- Mucosis B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | | | - Tianxu Sun
- College of Life Science, Jilin Agricultural University, Changchun 130118, China
| | - Tiejun Gu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China
| | - Yueshuang Song
- Changchun BCHT Biotechnology Co., Changchun 130012, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China
| | - Yongge Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| |
Collapse
|
24
|
Odutola A, Ota MOC, Antonio M, Ogundare EO, Saidu Y, Foster-Nyarko E, Owiafe PK, Ceesay F, Worwui A, Idoko OT, Owolabi O, Bojang A, Jarju S, Drammeh I, Kampmann B, Greenwood BM, Alderson M, Traskine M, Devos N, Schoonbroodt S, Swinnen K, Verlant V, Dobbelaere K, Borys D. Efficacy of a novel, protein-based pneumococcal vaccine against nasopharyngeal carriage of Streptococcus pneumoniae in infants: A phase 2, randomized, controlled, observer-blind study. Vaccine 2017; 35:2531-2542. [PMID: 28389097 DOI: 10.1016/j.vaccine.2017.03.071] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Conserved pneumococcal proteins are potential candidates for inclusion in vaccines against pneumococcal diseases. In the first part of a two-part study, an investigational vaccine (PHiD-CV/dPly/PhtD-30) containing 10 pneumococcal serotype-specific polysaccharide conjugates (10VT) combined with pneumolysin toxoid and pneumococcal histidine triad protein D (30μg each) was well tolerated by Gambian children. Part two, presented here, assessed the efficacy of two PHiD-CV/dPly/PhtD formulations against pneumococcal nasopharyngeal carriage (NPC) prevalence in infants. METHODS In this phase 2, randomized, controlled, observer-blind trial, healthy infants aged 8-10weeks, recruited from a peri-urban health center, were randomized (1:1:1:1:1:1) into six groups. Four groups received PHiD-CV/dPly/PhtD (10 or 30μg of each protein), PHiD-CV, or 13-valent pneumococcal conjugate vaccine at ages 2-3-4months (3+0 infant schedule) and two groups PHiD-CV/dPly/PhtD-30 or PHiD-CV at 2-4-9months (2+1 infant schedule). The primary objective was impact on non-10VT NPC at ages 5-9-12months. Secondary objectives included confirmatory analysis of protein dose superiority and safety/reactogenicity. Impact on pneumococcal NPC acquisition, bacterial load, and ply and phtD gene sequencing were explored. RESULTS 1200 infants were enrolled between June 2011 and May 2012. Prevalences of pneumococcal (60-67%) and non-10VT (55-61%) NPC were high at baseline. Across all post-vaccination time points, efficacy of PHiD-CV/dPly/PhtD-10 and PHiD-CV/dPly/PhtD-30 against non-10VT NPC (3+0 schedule) was 1.1% (95% CI -21.5, 19.5) and 2.1% (-20.3, 20.3), respectively; efficacy of PHiD-CV/dPly/PhtD-30 (2+1 schedule) was 0.5% (-22.1, 18.9) versus PHiD-CV. No differences were observed in pneumococcal NPC acquisition, clearance, or bacterial load. Both protein-based vaccines elicited immune responses to pneumococcal proteins. CONCLUSIONS In this high carriage prevalence setting, inclusion of pneumococcal proteins in the PHiD-CV/dPly/PhtD investigational vaccine had no impact on pneumococcal NPC in infants, regardless of protein dose or schedule. Future evaluations will assess its impact against pneumococcal disease endpoints. FUNDING PATH, GlaxoSmithKline Biologicals SA. ClinicalTrials.gov identifier NCT01262872.
Collapse
Affiliation(s)
- Aderonke Odutola
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Martin O C Ota
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Martin Antonio
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Ezra O Ogundare
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Yauba Saidu
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | | | - Patrick K Owiafe
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Fatima Ceesay
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Archibald Worwui
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Olubukola T Idoko
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Olumuyiwa Owolabi
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Abdoulie Bojang
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Sheikh Jarju
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Isatou Drammeh
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Beate Kampmann
- Medical Research Council Unit, Vaccines & Immunity Theme, Banjul, Gambia.
| | - Brian M Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Otitis-Prone Children Produce Functional Antibodies to Pneumolysin and Pneumococcal Polysaccharides. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00497-16. [PMID: 28031178 PMCID: PMC5339643 DOI: 10.1128/cvi.00497-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022]
Abstract
The pneumococcus is a major otitis media (OM) pathogen, but data are conflicting regarding whether otitis-prone children have impaired humoral immunity to pneumococcal antigens. We and others have shown that otitis-prone and healthy children have similar antibody titers to pneumococcal proteins and polysaccharides (vaccine and nonvaccine types); however, the quality of antibodies from otitis-prone children has not been investigated. Antibody function, rather than titer, is considered to be a better correlate of protection from pneumococcal disease. Therefore, we compared the capacities of antibodies from otitis-prone (cases) and healthy (controls) children to neutralize pneumolysin, the pneumococcal toxin currently in development as a vaccine antigen, and to opsonize pneumococcal vaccine and nonvaccine serotypes. A pneumolysin neutralization assay was conducted on cholesterol-depleted complement-inactivated sera from 165 cases and 61 controls. A multiplex opsonophagocytosis assay (MOPA) was conducted on sera from 20 cases and 20 controls. Neutralizing and opsonizing titers were calculated with antigen-specific IgG titers to determine antibody potency for pneumolysin, pneumococcal conjugate vaccine (PCV) polysaccharides, and non-PCV polysaccharides. There was no significant difference in antibody potencies between cases and controls for the antigens tested. Antipneumolysin neutralizing titers increased with the number of episodes of acute OM, but antibody potency did not. Pneumolysin antibody potency was lower in children colonized with pneumococci than in noncarriers, and this was significant for the otitis-prone group (P < 0.05). The production of functional antipneumococcal antibodies in otitis-prone children demonstrates that they respond to the current PCV and are likely to respond to pneumolysin-based vaccines as effectively as healthy children.
Collapse
|
26
|
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: 1.7] [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
|
27
|
Abstract
Streptococcus pneumoniae remains one of the most frequent bacterial causes of morbidity and mortality worldwide. National immunization programs implementing pneumococcal polysaccharide conjugate vaccines (PCVs) have successfully reduced rates of vaccine-type invasive disease and colonization both via direct effects in immunized children and, in some settings, indirect effects in unimmunized individuals. Limitations of the current PCV approach include the emergence of non-vaccine serotypes contributing to carriage and invasive disease in high-PCV coverage settings and the high cost of goods of PCVs which limits their accessibility in developing countries where the burden of disease remains highest. Furthermore, the distribution of serotypes causing disease varies geographically and includes more serotypes than are currently covered in a single PCV formulation. Researchers have long been exploring the potential of genetically conserved non-capsular pneumococcal antigens as vaccine candidates that might overcome such limitations. To better evaluate the rationale of such approaches, an understanding of the mechanisms of immunity to the various phases of pneumococcal infection is of paramount importance. Herein we will review the evolving understanding of both vaccine-induced and naturally acquired immunity to pneumococcal colonization and infection and discuss how this informs current approaches using serotype-independent pneumococcal vaccine candidates. We will then review the alternative vaccine candidates that have been or are currently under evaluation in clinical trials.
Collapse
Affiliation(s)
- Kristin Moffitt
- a Division of Infectious Diseases ; Department of Medicine; Boston Children's Hospital ; Boston , MA USA
| | - Richard Malley
- a Division of Infectious Diseases ; Department of Medicine; Boston Children's Hospital ; Boston , MA USA
| |
Collapse
|
28
|
Giebink GS, Bakaletz LO, Barenkamp SJ, Eskola J, Green B, Gu XX, Harada T, Heikkinen T, Karma P, Klein JO, Kurono Y, Mogi G, Murphy TF, Ogra PL, Patel JA, Suzuki M, Yamanaka N. 7. Vaccine. Ann Otol Rhinol Laryngol 2016. [DOI: 10.1177/00034894021110s310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
29
|
Abstract
INTRODUCTION The success of the vaccines available on the market has significantly increased interest in vaccine development. Areas covered: The main aim of this paper is to discuss the most important vaccines of pediatric interest that are currently being developed. New pneumococcal vaccines and vaccines against group B Streptococcus, Staphylococcus aureus and respiratory syncytial virus are analyzed in detail. Expert commentary: Advances in understanding human immunology, including human monoclonal antibody identification, sequencing technology, and the ability to solve atomic level structures of vaccine targets have provided tools to guide the rational design of future vaccines. It is likely that some of these vaccines will reach the market in the future and will thus partially contribute to the prevention of very severe diseases that significantly affect the morbidity and mortality of children. However, further studies in animals and several clinical trials in children must be performed before new vaccines become licensed.
Collapse
Affiliation(s)
- Susanna Esposito
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Nicola Principi
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
30
|
Daniels CC, Rogers PD, Shelton CM. A Review of Pneumococcal Vaccines: Current Polysaccharide Vaccine Recommendations and Future Protein Antigens. J Pediatr Pharmacol Ther 2016; 21:27-35. [PMID: 26997927 PMCID: PMC4778694 DOI: 10.5863/1551-6776-21.1.27] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This review describes development of currently available pneumococcal vaccines, provides summary tables of current pneumococcal vaccine recommendations in children and adults, and describes new potential vaccine antigens in the pipeline. Streptococcus pneumoniae, the bacteria responsible for pneumonia, otitis media, meningitis and bacteremia, remains a cause of morbidity and mortality in both children and adults. Introductions of unconjugated and conjugated pneumococcal polysaccharide vaccines have each reduced the rate of pneumococcal infections caused by the organism S. pneumoniae. The first vaccine developed, the 23-valent pneumococcal polysaccharide vaccine (PPSV23), protected adults and children older than 2 years of age against invasive disease caused by the 23 capsular serotypes contained in the vaccine. Because PPSV23 did not elicit a protective immune response in children younger than 2 years of age, the 7-valent pneumococcal conjugate vaccine (PCV7) containing seven of the most common serotypes from PPSV23 in pediatric invasive disease was developed for use in children younger than 2 years of age. The last vaccine to be developed, the 13-valent pneumococcal conjugate vaccine (PCV13), contains the seven serotypes in PCV7, five additional serotypes from PPSV23, and a new serotype not contained in PPSV23 or PCV7. Serotype replacement with virulent strains that are not contained in the polysaccharide vaccines has been observed after vaccine implementation and stresses the need for continued research into novel vaccine antigens. We describe eight potential protein antigens that are in the pipeline for new pneumococcal vaccines.
Collapse
Affiliation(s)
- Calvin C. Daniels
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - P. David Rogers
- Department of Clinical Pharmacy, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
- Center for Pediatric Pharmacokinetics and Therapeutics, Memphis, Tennessee
| | - Chasity M. Shelton
- Department of Clinical Pharmacy, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
31
|
Barnett TC, Cole JN, Rivera-Hernandez T, Henningham A, Paton JC, Nizet V, Walker MJ. Streptococcal toxins: role in pathogenesis and disease. Cell Microbiol 2015; 17:1721-41. [PMID: 26433203 DOI: 10.1111/cmi.12531] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/13/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022]
Abstract
Group A Streptococcus (Streptococcus pyogenes), group B Streptococcus (Streptococcus agalactiae) and Streptococcus pneumoniae (pneumococcus) are host-adapted bacterial pathogens among the leading infectious causes of human morbidity and mortality. These microbes and related members of the genus Streptococcus produce an array of toxins that act against human cells or tissues, resulting in impaired immune responses and subversion of host physiological processes to benefit the invading microorganism. This toxin repertoire includes haemolysins, proteases, superantigens and other agents that ultimately enhance colonization and survival within the host and promote dissemination of the pathogen.
Collapse
Affiliation(s)
- Timothy C Barnett
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Jason N Cole
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Tania Rivera-Hernandez
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Anna Henningham
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Mark J Walker
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
32
|
Diversity of Pneumolysin and Pneumococcal Histidine Triad Protein D of Streptococcus pneumoniae Isolated from Invasive Diseases in Korean Children. PLoS One 2015; 10:e0134055. [PMID: 26252211 PMCID: PMC4529296 DOI: 10.1371/journal.pone.0134055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/04/2015] [Indexed: 01/07/2023] Open
Abstract
Pneumolysin (Ply) and pneumococcal histidine triad protein D (PhtD) are candidate proteins for a next-generation pneumococcal vaccine. We aimed to analyze the genetic diversity and antigenic heterogeneity of Ply and PhtD for 173 pneumococci isolated from invasive diseases in Korean children. Allele was designated based on the variation of amino acid sequence. Antigenicity was predicted by the amino acid hydrophobicity of the region. There were seven and 39 allele types for the ply and phtD genes, respectively. The nucleotide sequence identity was 97.2%-99.9% for ply and 91.4%-98.0% for phtD gene. Only minor variations in hydrophobicity were noted among the antigenicity plots of Ply and PhtD. Overall, the allele types of the ply and phtD genes were remarkably homogeneous, and the antigenic diversity of the corresponding proteins was very limited. The Ply and PhtD could be useful antigens for universal pneumococcal vaccines.
Collapse
|
33
|
Multivalent Pneumococcal Protein Vaccines Comprising Pneumolysoid with Epitopes/Fragments of CbpA and/or PspA Elicit Strong and Broad Protection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:1079-89. [PMID: 26245351 DOI: 10.1128/cvi.00293-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/28/2015] [Indexed: 12/11/2022]
Abstract
Immunization with the pneumococcal proteins pneumolysin (Ply), choline binding protein A (CbpA), or pneumococcal surface protein A (PspA) elicits protective responses against invasive pneumococcal disease in animal models. In this study, we used different mouse models to test the efficacy of a variety of multivalent protein-based vaccines that comprised various combinations of full-length or peptide regions of the immunogens Ply, CbpA, or PspA: Ply toxoid with the L460D substitution (referred to herein as L460D); L460D fused with protective peptide epitopes from CbpA (YPT-L460D-NEEK [YLN]); L460D fused with the CD2 peptide containing the proline-rich region (PRR) of PspA (CD2-L460D); a combination of L460D and H70 (L460D+H70), a slightly larger PspA-derived peptide containing the PRR and the SM1 region; H70+YLN; and other combinations. Each mouse was immunized either intraperitoneally (i.p.) or subcutaneously (s.c.) with three doses (at 2-week intervals) of the various antigen combinations in alum adjuvant and then challenged in mouse models featuring different infection routes with multiple Streptococcus pneumoniae strains. In the i.p. infection sepsis model, H70+YLN consistently provided significant protection against three different challenge strains (serotypes 1, 2, and 6A); the CD2+YLN and H70+L460D combinations also elicited significant protection. Protection against intravenous (i.v.) sepsis (type 3 and 6A challenge strains) was largely dependent on PspA-derived antigen components, and the most protection was elicited by H70 with or without L460D or YLN. In a type 4 intratracheal (i.t.) challenge model that results in progression to meningitis, antigen combinations that contained YLN elicited the strongest protection. Thus, the trivalent antigen combination of H70+YLN elicited the strongest and broadest protection in diverse pneumococcal challenge models.
Collapse
|
34
|
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: 9.1] [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
|
35
|
Identification of proteins in Streptococcus pneumoniae by reverse vaccinology and genetic diversity of these proteins in clinical isolates. Appl Biochem Biotechnol 2014; 175:2124-65. [PMID: 25448632 DOI: 10.1007/s12010-014-1375-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 11/10/2014] [Indexed: 10/24/2022]
Abstract
Streptococcus pneumoniae is a major cause of morbidity and mortality worldwide. Virulence-associated proteins common and conserved among all capsular types now represent the best strategy to combat pneumococcal infections. Our aim was to identify conserved targets in pneumococci that showed positive prediction for lipoprotein and extracellular subcellular location using bioinformatics programs and verify the distribution and the degree of conservation of these targets in pneumococci. These targets can be considered potential vaccine candidate to be evaluated in the future. A set of 13 targets were analyzed and confirmed the presence in all pneumococci tested. These 13 genes were highly conserved showing around >96 % of amino acid and nucleotide identity, but they were also present and show high identity in the closely related species Streptococcus mitis, Streptococcus oralis, and Streptococcus pseudopneumoniae. S. oralis clusters away from S. pneumoniae, while S. pseudopneumoniae and S. mitis cluster closer. The divergence between the selected targets was too small to be observed consistently in phylogenetic groups between the analyzed genomes of S. pneumoniae. The proteins analyzed fulfill two of the initial criteria of a vaccine candidate: targets are present in a variety of different pneumococci strains including different serotypes and are conserved among the samples evaluated.
Collapse
|
36
|
Leroux-Roels G, Maes C, De Boever F, Traskine M, Rüggeberg JU, Borys D. Safety, reactogenicity and immunogenicity of a novel pneumococcal protein-based vaccine in adults: A phase I/II randomized clinical study. Vaccine 2014; 32:6838-46. [DOI: 10.1016/j.vaccine.2014.02.052] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/20/2014] [Accepted: 02/10/2014] [Indexed: 10/25/2022]
|
37
|
Human antibodies to PhtD, PcpA, and Ply reduce adherence to human lung epithelial cells and murine nasopharyngeal colonization by Streptococcus pneumoniae. Infect Immun 2014; 82:5069-75. [PMID: 25245804 DOI: 10.1128/iai.02124-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Streptococcus pneumoniae adherence to human epithelial cells (HECs) is the first step in pathogenesis leading to infections. We sought to determine the role of human antibodies against S. pneumoniae protein vaccine candidates PhtD, PcpA, and Ply in preventing adherence to lung HECs in vitro and mouse nasopharyngeal (NP) colonization in vivo. Human anti-PhtD, -PcpA, and -Ply antibodies were purified and Fab fragments generated. Fabs were used to test inhibition of adherence of TIGR4 and nonencapsulated strain RX1 to A549 lung HECs. The roles of individual proteins in adherence were tested using isogenic mutants of strain TIGR4. Anti-PhtD, -PcpA, and -Ply human antibodies were assessed for their ability to inhibit NP colonization in vivo by passive transfer of human antibody in a murine model. Human antibodies generated against PhtD and PcpA caused a decrease in adherence to A549 cells (P < 0.05). Anti-PhtD but not anti-PcpA antibodies showed a protective role against mouse NP colonization. To our surprise, anti-Ply antibodies also caused a significant (P < 0.05) reduction in S. pneumoniae colonization. Our results support the potential of PhtD, PcpA, and Ply protein vaccine candidates as alternatives to conjugate vaccines to prevent non-serotype-specific S. pneumoniae colonization and invasive infection.
Collapse
|
38
|
Smith CM, Sandrini S, Datta S, Freestone P, Shafeeq S, Radhakrishnan P, Williams G, Glenn SM, Kuipers OP, Hirst RA, Easton AJ, Andrew PW, O'Callaghan C. Respiratory syncytial virus increases the virulence of Streptococcus pneumoniae by binding to penicillin binding protein 1a. A new paradigm in respiratory infection. Am J Respir Crit Care Med 2014; 190:196-207. [PMID: 24941423 DOI: 10.1164/rccm.201311-2110oc] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Respiratory syncytial virus (RSV) and Streptococcus pneumoniae are major respiratory pathogens. Coinfection with RSV and S. pneumoniae is associated with severe and often fatal pneumonia but the molecular basis for this remains unclear. OBJECTIVES To determine if interaction between RSV and pneumococci enhances pneumococcal virulence. METHODS We used confocal microscopy and Western blot to identify the receptors involved in direct binding of RSV and pneumococci, the effects of which were studied in both in vivo and in vitro models of infection. Human ciliated respiratory epithelial cell cultures were infected with RSV for 72 hours and then challenged with pneumococci. Pneumococci were collected after 2 hours exposure and changes in gene expression determined using quantitative real-time polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS Following incubation with RSV or purified G protein, pneumococci demonstrated a significant increase in the inflammatory response and bacterial adherence to human ciliated epithelial cultures and markedly increased virulence in a pneumonia model in mice. This was associated with extensive changes in the pneumococcal transcriptome and significant up-regulation in the expression of key pneumococcal virulence genes, including the gene for the pneumococcal toxin, pneumolysin. We show that mechanistically this is caused by RSV G glycoprotein binding penicillin binding protein 1a. CONCLUSIONS The direct interaction between a respiratory virus protein and the pneumococcus resulting in increased bacterial virulence and worsening disease outcome is a new paradigm in respiratory infection.
Collapse
Affiliation(s)
- Claire M Smith
- 1 Respiratory, Critical Care and Anaesthesia, University College London, Institute of Child Health, Great Ormond Street Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Harvey RM, Hughes CE, Paton AW, Trappetti C, Tweten RK, Paton JC. The impact of pneumolysin on the macrophage response to Streptococcus pneumoniae is strain-dependent. PLoS One 2014; 9:e103625. [PMID: 25105894 PMCID: PMC4126675 DOI: 10.1371/journal.pone.0103625] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/28/2014] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pneumoniae is the world's leading cause of pneumonia, bacteremia, meningitis and otitis media. A major pneumococcal virulence factor is the cholesterol-dependent cytolysin, which has the defining property of forming pores in cholesterol-containing membranes. In recent times a clinically significant and internationally successful serotype 1 ST306 clone has been found to express a non-cytolytic variant of Ply (Ply306). However, while the pneumococcus is a naturally transformable organism, strains of the ST306 clonal group have to date been virtually impossible to transform, severely restricting efforts to understand the role of non-cytolytic Ply in the success of this clone. In this study isogenic Ply mutants were constructed in the D39 background and for the first time in the ST306 background (A0229467) to enable direct comparisons between Ply variants for their impact on the immune response in a macrophage-like cell line. Strains that expressed cytolytic Ply were found to induce a significant increase in IL-1β release from macrophage-like cells compared to the non-cytolytic and Ply-deficient strains in a background-independent manner, confirming the requirement for pore formation in the Ply-dependent activation of the NLRP3 inflammasome. However, cytolytic activity in the D39 background was found to induce increased expression of the genes encoding GM-CSF (CSF2), p19 subunit of IL-23 (IL23A) and IFNβ (IFNB1) compared to non-cytolytic and Ply-deficient D39 mutants, but had no effect in the A0229467 background. The impact of Ply on the immune response to the pneumococcus is highly dependent on the strain background, thus emphasising the importance of the interaction between specific virulence factors and other components of the genetic background of this organism.
Collapse
Affiliation(s)
- Richard M. Harvey
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Catherine E. Hughes
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Adrienne W. Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Claudia Trappetti
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Rodney K. Tweten
- Department of Microbiology & Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - James C. Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
- * E-mail:
| |
Collapse
|
40
|
Lu J, Sun T, Hou H, Xu M, Gu T, Dong Y, Wang D, Chen P, Wu C, Liang C, Sun S, Jiang C, Kong W, Wu Y. Detoxified pneumolysin derivative plym2 directly protects against pneumococcal infection via induction of inflammatory cytokines. Immunol Invest 2014; 43:717-26. [DOI: 10.3109/08820139.2014.930478] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Prymula R, Pazdiora P, Traskine M, Rüggeberg JU, Borys D. Safety and immunogenicity of an investigational vaccine containing two common pneumococcal proteins in toddlers: A phase II randomized clinical trial. Vaccine 2014; 32:3025-34. [DOI: 10.1016/j.vaccine.2014.03.066] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/28/2014] [Accepted: 03/17/2014] [Indexed: 11/28/2022]
|
42
|
Pauksens K, Nilsson AC, Caubet M, Pascal TG, Van Belle P, Poolman JT, Vandepapelière PG, Verlant V, Vink PE. Randomized controlled study of the safety and immunogenicity of pneumococcal vaccine formulations containing PhtD and detoxified pneumolysin with alum or adjuvant system AS02V in elderly adults. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:651-60. [PMID: 24599529 PMCID: PMC4018883 DOI: 10.1128/cvi.00807-13] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/24/2014] [Indexed: 12/20/2022]
Abstract
Six vaccine formulations containing AS02V or alum (aluminum phosphate [AlPO4]) adjuvant with pneumococcal proteins, pneumococcal histidine triad D (PhtD), and/or detoxified pneumolysin (dPly), either as a polysaccharide carrier in an 8-valent pneumococcal conjugate vaccine (8PCV) or as free (unconjugated) proteins, were evaluated in adults -65 to 85 years of age. In this phase I observer-blind study, 167 healthy subjects were randomized to receive two doses (days 0 and 60) of 10 or 30 μg PhtD-dPly plus AS02V or alum, 8PCV plus AS02V or alum, or one dose (day 0) of 23-valent polysaccharide pneumococcal vaccine (23PPV) as a control (placebo on day 60). The safety, reactogenicity, and antibody-specific responses to these vaccines were evaluated. No vaccine-related serious adverse events were reported. The incidences of solicited local and specific general (fatigue and myalgia) symptoms tended to be higher in the AS02V groups than in other groups. Anti-PhtD and anti-Ply antibody responses were observed in all groups except the control group. One month post-dose 2, the anti-PhtD and anti-Ply antibody geometric mean concentrations tended to be higher with AS02V than with alum, higher with a dose of 30 μg than with 10 μg for PhtD-dPly and higher with 30-μg PhtD-dPly formulations than with conjugated PhtD and dPly (8PCV) formulations. Functional antibody responses, measured by an opsonophagocytic activity assay, tended to be higher with 8PCV than with 23PPV. In conclusion, vaccine formulations containing free or conjugated PhtD and dPly had acceptable reactogenicity and safety profiles in elderly adults. Immune responses were enhanced with an AS02V-adjuvanted formulation containing free 30-μg PhtD-dPly compared to those with alum adjuvant and conjugated proteins. (This study has been registered at ClinicalTrials.gov under registration no. NCT00756067.).
Collapse
Affiliation(s)
- Karlis Pauksens
- Department of Infectious Diseases, Uppsala University Hospital, Akademiska Sjukhuset, Uppsala, Sweden
| | - Anna C. Nilsson
- Department of Clinical Sciences, Malmö Infectious Disease Research Unit, Lund University, Lund, Sweden
| | | | | | | | | | | | | | - Peter E. Vink
- GlaxoSmithKline Vaccines, King of Prussia, Pennsylvania, USA
| |
Collapse
|
43
|
Immune responses to pneumococcal pilus RrgA and RrgB antigens and their relationship with pneumococcal carriage in humans. J Infect 2014; 68:562-71. [PMID: 24509142 DOI: 10.1016/j.jinf.2014.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/17/2014] [Accepted: 01/30/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Pneumococcal pilus antigens are shown to be important in pneumococcal pathogenesis and induce protective immunity in animal studies, but data in humans are limited. We aimed to investigate serum and mucosal immune responses to pilus-1 proteins (RrgA and RrgB) and their relationship with pneumococcal carriage in humans. METHODS Serum and salivary antibodies to RrgA and RrgB in children and adults were analysed by ELISA and immunoblotting. Induction of B cell antibody responses to RrgA and RrgB in nasopharynx-associated lymphoid tissue was studied by ELISpot assay following stimulation with pneumococcal culture supernatants containing pilus proteins. RESULTS Significant levels of serum anti-RrgA and -RrgB antibodies were observed, and anti-RrgA antibody appeared to develop earlier in childhood. Importantly, anti-RrgA IgG titres in both serum and saliva were shown to be higher in culture-negative children than in those who were culture-positive for Streptococcus pneumoniae. Stimulation of adenotonsillar cells with pneumococcal culture supernatant induced significant RrgA- and RrgB-specific antibody secreting cells and antibody production. CONCLUSIONS Pneumococcal pilus antigens, particularly RrgA, seem to induce significant serum and mucosal antibody responses that may contribute to natural immunity against pneumococcal carriage in children.
Collapse
|
44
|
Abstract
Cholesterol dependent cytolysins are important in the ability of some bacteria to cause disease in man and animals. Pneumolysin (PLY) plays a key role in the diseases caused by Streptococcus pneumoniae (the pneumococcus). This chapter describes the role of PLY in some of the key process in disease. These include induction of cell death by pore formation and toxin-induced apoptosis as well as more subtle effects on gene expression of host cells including epigenetic effects of the toxin. The use of bacterial mutants that either do not express the toxin or express altered versions in biological systems is described. Use of isolated tissue and whole animal systems to dissect the structure/function relationships of the toxin as well as the role played by different activities in the pathogenesis of infection are described. The role of PLY in meningitis and the associated deafness is discussed as well as the role of the toxin in promoting increased lung permeability and inflammation during pneumococcal pneumonia. Different clinical strains of the pneumococcus produce different forms of PLY and the impact of this on disease caused by these strains is discussed. Finally, the impact of this knowledge on the development of treatment and prevention strategies for pneumococcal disease is discussed.
Collapse
|
45
|
Rosch JW, Iverson AR, Humann J, Mann B, Gao G, Vogel P, Mina M, Murrah KA, Perez AC, Edward Swords W, Tuomanen EI, McCullers JA. A live-attenuated pneumococcal vaccine elicits CD4+ T-cell dependent class switching and provides serotype independent protection against acute otitis media. EMBO Mol Med 2014; 6:141-54. [PMID: 24408968 PMCID: PMC3936495 DOI: 10.1002/emmm.201202150] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 09/06/2013] [Accepted: 09/23/2013] [Indexed: 01/19/2023] Open
Abstract
Acute otitis media (AOM) caused by Streptococcus pneumoniae remains one of the most common infectious diseases worldwide despite widespread vaccination. A major limitation of the currently licensed pneumococcal vaccines is the lack of efficacy against mucosal disease manifestations such as AOM, acute bacterial sinusitis and pneumonia. We sought to generate a novel class of live vaccines that (1) retain all major antigenic virulence proteins yet are fully attenuated and (2) protect against otitis media. A live vaccine candidate based on deletion of the signal recognition pathway component ftsY induced potent, serotype-independent protection against otitis media, sinusitis, pneumonia and invasive pneumococcal disease. Protection was maintained in animals coinfected with influenza virus, but was lost if mice were depleted of CD4(+) T cells at the time of vaccination. The live vaccine induced a strong serum IgG2a and IgG2b response that correlated with CD4(+) T-cell mediated class switching. Deletion of genes required for microbial adaptation to the host environment is a novel live attenuated vaccine strategy yielding the first experimental vaccine effective against pneumococcal otitis media.
Collapse
Affiliation(s)
- Jason W Rosch
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Amy R Iverson
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Jessica Humann
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Beth Mann
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Geli Gao
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Michael Mina
- Emory University School of MedicineAtlanta, GA, USA
| | - Kyle A Murrah
- Department of Microbiology and Immunology, Wake Forest School of MedicineWinston-Salem, NC, USA
| | - Antonia C Perez
- Department of Microbiology and Immunology, Wake Forest School of MedicineWinston-Salem, NC, USA
| | - W Edward Swords
- Department of Microbiology and Immunology, Wake Forest School of MedicineWinston-Salem, NC, USA
| | - Elaine I Tuomanen
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| | - Jonathan A McCullers
- Department of Infectious Diseases, St. Jude Children's Research HospitalMemphis, TN, USA
| |
Collapse
|
46
|
Kirkham LAS, Corscadden KJ, Wiertsema SP, Currie AJ, Richmond PC. A practical method for preparation of pneumococcal and nontypeable Haemophilus influenzae inocula that preserves viability and immunostimulatory activity. BMC Res Notes 2013; 6:522. [PMID: 24321049 PMCID: PMC3867214 DOI: 10.1186/1756-0500-6-522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 12/04/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Convenience is a major reason for using killed preparations of bacteria to investigate host-pathogen interactions, however, host responses to such preparations can result in different outcomes when compared to live bacterial stimulation. We investigated whether cryopreservation of Streptococcus pneumoniae and nontypeable Haemophilus influenzae (NTHi) permitted investigation of host responses to infection without the complications of working with freshly prepared live bacteria on the day of experimental challenge. FINDINGS S. pneumoniae and NTHi retained >90% viability following cryopreservation in fetal calf serum for at least 8 weeks. Host responses to live, cryopreserved (1 week and 4 weeks), heat-killed or ethanol-killed S. pneumoniae and NTHi were assessed by measuring cytokine release from stimulated peripheral blood mononuclear cells (PBMCs). We found that cryopreserved bacteria, in contrast to heat-killed and ethanol-killed preparations, resulted in comparable levels of inflammatory cytokine release from PBMCs when compared with fresh live bacterial cultures. CONCLUSION Cryopreservation of S. pneumoniae and NTHi does not alter the immunostimulatory properties of these species thereby enabling reproducible and biologically relevant analysis of host responses to infection. This method also facilitates the analysis of multiple strains on the same day and allows predetermination of culture purity and challenge dose.
Collapse
Affiliation(s)
- Lea-Ann S Kirkham
- School of Paediatrics and Child Health, The University of Western Australia, 35 Stirling Highway, Perth, WA 6009, Australia.
| | | | | | | | | |
Collapse
|
47
|
Safety, immunogenicity, and antibody persistence following an investigational Streptococcus pneumoniae and Haemophilus influenzae triple-protein vaccine in a phase 1 randomized controlled study in healthy adults. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 21:56-65. [PMID: 24173029 DOI: 10.1128/cvi.00430-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We investigated a protein-based nontypeable Haemophilus influenzae (NTHi) and pneumococcal (HiP) vaccine containing pneumococcal histidine triad D (PhtD), detoxified pneumolysin (dPly), and NTHi protein D (PD) in adults. In a phase I study, 40 healthy 18- to 40-year-old subjects were randomized (2:2:1) to receive two HiP doses administered 60 days apart, with or without AS03 adjuvant (HiP-AS and HiP groups, respectively), or Engerix B (GlaxoSmithKline, Belgium) as a control. Safety, antibodies, and antigen-specific CD4(+) T-cell immune responses were assessed before and until 480 days after vaccination. No serious adverse events were reported, and no subject withdrew due to an adverse event. Local and systemic symptoms were reported more frequently in the HiP-AS group than in the other two groups. The frequency and intensity of local and systemic symptoms appeared to increase after the second dose of HiP-AS or HiP but not Engerix B. Antibody geometric mean concentrations (GMCs) for PhtD, dPly, and PD increased after each dose of HiP-AS or HiP, with higher GMCs being observed in the HiP-AS group (statistically significant for anti-PD after dose 1 and anti-Ply after dose 2). GMCs remained higher at day 420 than prior to vaccination in both the HiP-AS and HiP groups. Antigen-specific CD4(+) T cells increased after each dose but were unmeasurable by day 480. Two doses of an investigational PhtD-dPly-PD protein vaccine induced humoral immunity and antigen-specific CD4(+) T-cell responses after each dose, with generally higher responses when the vaccine was administered with AS03. HiP combined with AS03 appeared to be more reactogenic than the antigens alone. (This study has been registered at ClinicalTrials.gov under registration no. NCT00814489.).
Collapse
|
48
|
Miyaji EN, Oliveira MLS, Carvalho E, Ho PL. Serotype-independent pneumococcal vaccines. Cell Mol Life Sci 2013; 70:3303-26. [PMID: 23269437 PMCID: PMC11113425 DOI: 10.1007/s00018-012-1234-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/01/2012] [Accepted: 12/04/2012] [Indexed: 12/14/2022]
Abstract
Streptococcus pneumoniae remains an important cause of disease with high mortality and morbidity, especially in children and in the elderly. The widespread use of the polysaccharide conjugate vaccines in some countries has led to a significant decrease in invasive disease caused by vaccine serotypes, but an increase in disease caused by non-vaccine serotypes has impacted on the overall efficacy of these vaccines on pneumococcal disease. The obvious solution to overcome such shortcomings would be the development of new formulations that provide serotype-independent immunity. This review focuses on the most promising approaches, including protein antigens, whole cell pneumococcal vaccines, and recombinant bacteria expressing pneumococcal antigens. The protective capacity of these vaccine candidates against the different stages of pneumococcal infection, including colonization, mucosal disease, and invasive disease in animal models is reviewed. Some of the human trials that have already been performed or that are currently ongoing are presented. Finally, the feasibility and the possible shortcomings of these candidates in relation to an ideal vaccine against pneumococcal infections are discussed.
Collapse
Affiliation(s)
- Eliane Namie Miyaji
- Centro de Biotecnologia, Instituto Butantan, Av Vital Brasil 1500, São Paulo, SP 05503-900 Brazil
| | | | - Eneas Carvalho
- Centro de Biotecnologia, Instituto Butantan, Av Vital Brasil 1500, São Paulo, SP 05503-900 Brazil
| | - Paulo Lee Ho
- Centro de Biotecnologia, Instituto Butantan, Av Vital Brasil 1500, São Paulo, SP 05503-900 Brazil
| |
Collapse
|
49
|
Olaya-Abril A, Jiménez-Munguía I, Gómez-Gascón L, Obando I, Rodríguez-Ortega MJ. Identification of potential new protein vaccine candidates through pan-surfomic analysis of pneumococcal clinical isolates from adults. PLoS One 2013; 8:e70365. [PMID: 23894641 PMCID: PMC3720901 DOI: 10.1371/journal.pone.0070365] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/17/2013] [Indexed: 11/23/2022] Open
Abstract
Purified polysaccharide and conjugate vaccines are widely used for preventing infections in adults and in children against the Gram-positive bacterium Streptococcus pneumoniae, a pathogen responsible for high morbidity and mortality rates, especially in developing countries. However, these polysaccharide-based vaccines have some important limitations, such as being serotype-dependent, being subjected to losing efficacy because of serotype replacement and high manufacturing complexity and cost. It is expected that protein-based vaccines will overcome these issues by conferring a broad coverage independent of serotype and lowering production costs. In this study, we have applied the “shaving” proteomic approach, consisting of the LC/MS/MS analysis of peptides generated by protease treatment of live cells, to a collection of 16 pneumococcal clinical isolates from adults, representing the most prevalent strains circulating in Spain during the last years. The set of unique proteins identified in all the isolates, called “pan-surfome”, consisted of 254 proteins, which included most of the protective protein antigens reported so far. In search of new candidates with vaccine potential, we identified 32 that were present in at least 50% of the clinical isolates analyzed. We selected four of them (Spr0012, Spr0328, Spr0561 and SP670_2141), whose protection capacity has not yet been tested, for assaying immunogenicity in human sera. All of them induced the production of IgM antibodies in infected patients, thus indicating that they could enter the pipeline for vaccine studies. The pan-surfomic approach shows its utility in the discovery of new proteins that can elicit protection against infectious microorganisms.
Collapse
Affiliation(s)
- Alfonso Olaya-Abril
- Departamento de Bioquímica y Biología Molecular, Universidad de Córdoba; Campus de Excelencia Internacional CeiA3; Hospital Universitario Reina Sofía, Córdoba; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba; and Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Jiménez-Munguía
- Departamento de Bioquímica y Biología Molecular, Universidad de Córdoba; Campus de Excelencia Internacional CeiA3; Hospital Universitario Reina Sofía, Córdoba; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba; and Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Ignacio Obando
- Sección de Enfermedades Infecciosas Pediátricas e Inmunopatología, Hospital Universitario Infantil Virgen del Rocío, Sevilla, Spain
| | - Manuel J. Rodríguez-Ortega
- Departamento de Bioquímica y Biología Molecular, Universidad de Córdoba; Campus de Excelencia Internacional CeiA3; Hospital Universitario Reina Sofía, Córdoba; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba; and Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
50
|
Taylor SD, Sanders ME, Tullos NA, Stray SJ, Norcross EW, McDaniel LS, Marquart ME. The cholesterol-dependent cytolysin pneumolysin from Streptococcus pneumoniae binds to lipid raft microdomains in human corneal epithelial cells. PLoS One 2013; 8:e61300. [PMID: 23577214 PMCID: PMC3618221 DOI: 10.1371/journal.pone.0061300] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 03/11/2013] [Indexed: 11/22/2022] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is an opportunistic bacterial pathogen responsible for causing several human diseases including pneumonia, meningitis, and otitis media. Pneumococcus is also a major cause of human ocular infections and is commonly isolated in cases of bacterial keratitis, an infection of the cornea. The ocular pathology that occurs during pneumococcal keratitis is partly due to the actions of pneumolysin (Ply), a cholesterol-dependent cytolysin produced by pneumococcus. The lytic mechanism of Ply is a three step process beginning with surface binding to cholesterol. Multiple Ply monomers then oligomerize to form a prepore. The prepore then undergoes a conformational change that creates a large pore in the host cell membrane, resulting in cell lysis. We engineered a collection of single amino acid substitution mutants at residues (A370, A406, W433, and L460) that are crucial to the progression of the lytic mechanism and determined the effects that these mutations had on lytic function. Both PlyWT and the mutant Ply molecules (PlyA370G, PlyA370E, PlyA406G, PlyA406E, PlyW433G, PlyW433E, PlyW433F, PlyL460G, and PlyL460E) were able to bind to the surface of human corneal epithelial cells (HCECs) with similar efficiency. Additionally, PlyWT localized to cholesterol-rich microdomains on the HCEC surface, however, only one mutant (PlyA370G) was able to duplicate this behavior. Four of the 9 mutant Ply molecules (PlyA370E, PlyW433G, PlyW433E, and PlyL460E) were deficient in oligomer formation. Lastly, all of the mutant Ply molecules, except PlyA370G, exhibited significantly impaired lytic activity on HCECs. The other 8 mutants all experienced a reduction in lytic activity, but 4 of the 8 retained the ability to oligomerize. A thorough understanding of the molecular interactions that occur between Ply and the target cell, could lead to targeted treatments aimed to reduce the pathology observed during pneumococcal keratitis.
Collapse
Affiliation(s)
- Sidney D. Taylor
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Melissa E. Sanders
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Nathan A. Tullos
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Stephen J. Stray
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Erin W. Norcross
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Larry S. McDaniel
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Mary E. Marquart
- Department of Microbiology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| |
Collapse
|