1
|
Vega LA, Sanson-Iglesias M, Mukherjee P, Buchan KD, Morrison G, Hohlt AE, Flores AR. LiaR-dependent gene expression contributes to antimicrobial responses in group A Streptococcus. Antimicrob Agents Chemother 2024; 68:e0049624. [PMID: 39535201 PMCID: PMC11619527 DOI: 10.1128/aac.00496-24] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
The ability to sense and respond to host defenses is essential for pathogen survival. Some mechanisms involve two-component systems (TCSs) that respond to host molecules, such as antimicrobial peptides (AMPs), and activate specific gene regulatory pathways to aid in survival. Alongside TCSs, bacteria coordinate cell division proteins, chaperones, cell wall sortases, and secretory translocons at discrete locations within the cytoplasmic membrane, referred to as functional membrane microdomains (FMMs). In group A Streptococcus (GAS), the FMM or "ExPortal" coordinates protein secretion, cell wall synthesis, and sensing of AMP-mediated cell envelope stress via the LiaFSR three-component system. Previously, we showed that GAS exposure to a subset of AMPs (α-defensins) activates the LiaFSR system by disrupting LiaF and LiaS co-localization in the ExPortal, leading to increased LiaR phosphorylation, expression of the transcriptional regulator SpxA2, and altered GAS virulence gene expression. The mechanisms by which LiaFSR integrates cell envelope stress with responses to AMP activity and virulence are not fully elucidated. Here, we show the LiaFSR regulon is comprised of genes encoding SpxA2 and three membrane-associated proteins: a PspC domain-containing protein (PCP), the lipoteichoic acid-modifying protein LafB, and the membrane protein insertase YidC2. Our data support that phosphorylated LiaR induces transcription of these genes via a conserved operator, whose disruption attenuates GAS virulence and increases susceptibility to AMPs in a manner primarily dependent on differential expression of SpxA2. Our work expands our understanding of the LiaFSR regulatory network in GAS and identifies targets for further investigation of mechanisms of cell envelope stress tolerance contributing to GAS pathogenesis.
Collapse
Affiliation(s)
- Luis Alberto Vega
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| | - Misú Sanson-Iglesias
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| | - Piyali Mukherjee
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| | - Kyle D. Buchan
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| | - Gretchen Morrison
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| | - Anne E. Hohlt
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| | - Anthony R. Flores
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston and Children’s Memorial Hermann Hospital, Houston, Texas, USA
| |
Collapse
|
2
|
Davis SE, Hart MT, Braza RED, Perry AA, Vega LA, Le Breton YS, McIver KS. The PdxR-PdxKU locus involved in vitamin B 6 salvage is important for group A streptococcal resistance to neutrophil killing and survival in human blood. Microbiol Spectr 2024; 12:e0160924. [PMID: 39530679 PMCID: PMC11619246 DOI: 10.1128/spectrum.01609-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Streptococcus pyogenes (Group A Streptococcus, GAS) is a Gram-positive bacterium that inflicts both superficial and life-threatening diseases on its human host. Analysis of fitness using a transposon mutant library revealed that genes predicted to be involved in vitamin B6 acquisition are associated with fitness in whole human blood. Vitamin B6 is essential for all life and is important for many cellular functions. In several streptococcal species, it has been shown that mutants in B6 acquisition exhibited reduced virulence phenotypes and were attenuated during infection. In GAS, B6 acquisition is believed to be controlled by the pdxR-pdxKU locus, where PdxR is a positive regulator of pdxKU, which encodes for a B6-substrate kinase and permease, respectively. Mutants in the regulator (ΔpdxR) and salvage machinery (ΔpdxKU) both exhibited modest growth defects when grown in oxygenated conditions with limited vitamin B6 precursors. ∆pdxR and ∆pdxKU mutants also exhibited an impaired ability to survive when challenged with whole human or mouse blood. This defect was characterized by reduced survival in the presence of human neutrophil-like HL60s, primary polymorphonuclear leukocytes, and antimicrobial peptide LL-37. Promoter analysis showed that PdxR is an autoregulator and activated pdxKU in the absence of B6. Interestingly, ∆pdxR and ∆pdxKU mutants were not attenuated in mouse models of infection, suggesting a species-specific impact on virulence. Overall, it appears that pdxR-pdxKU is associated with GAS vitamin B6 metabolism as well as pathogen survival during encounters with the human innate immune system.IMPORTANCEBacterial pathogens such as Streptococcus pyogenes (Group A Streptococcus, GAS) must be able to obtain needed nutrients in their human host. Vitamin B6 or pyridoxal 5' phosphate is essential for all life and is important for many cellular functions. In other streptococcal pathogens, B6 acquisition has been shown to be important for their ability to cause disease. Here, we show that loss of the putative vitamin B6 salvage pathway locus pdxR-pdxKU affects GAS pathogenesis when encountering innate immune responses from phagocytic neutrophils and antimicrobial peptides within the host. pdxR-pdxKU may contribute to oxygen tolerance through B6; however, there appear to be other mechanisms for salvaging vitamin B6. Overall, pdxR-pdxKU is associated with GAS resistance to the human innate immune response and oxygen tolerance and contributes modestly to B6 metabolism.
Collapse
Affiliation(s)
- Sarah E. Davis
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Meaghan T. Hart
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Rezia Era D. Braza
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Aolani A. Perry
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Luis A. Vega
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Yoann S. Le Breton
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kevin S. McIver
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
3
|
Anandan V, Bao L, Zhu Z, Bradley J, Assi VF, Chavda H, Kitten T, Xu P. A novel infective endocarditis virulence factor related to multiple functions for bacterial survival in blood was discovered in Streptococcus sanguinis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601854. [PMID: 39005390 PMCID: PMC11244957 DOI: 10.1101/2024.07.03.601854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
We identified the role of a conserved hypothetical protein (SSA_0451) in S. sanguinis that is involved in the virulence of infective endocarditis. An in vitro whole blood killing assay and rabbit endocarditis model studies revealed that the SSA_0451 mutant (ΔSSA_0451) was significantly less virulent than the wild-type (SK36) and its complementation mutant (ΔSSA_0451C). The mechanism underlying the SSA_0451 mutant's reduced virulence in infective endocarditis was evidentially linked to oxidative stress and environmental stress. The genes related to the survival of S. sanguinis in an oxidative stress environment were downregulated in ΔSSA_0451, which affected its survival in blood. Our findings suggest that SSA_0451 is a novel IE virulence factor and a new target for drug discovery against IE.
Collapse
Affiliation(s)
- Vysakh Anandan
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Liang Bao
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Zan Zhu
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Jennifer Bradley
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Valery-Francine Assi
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Henna Chavda
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Todd Kitten
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
| | - Ping Xu
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
- Center for Biological Data Science, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
4
|
Morita K, Moriwaki T, Habe S, Taniguchi-Ikeda M, Hasegawa T, Minato Y, Aoi T, Maruyama T. Molecular Aggregation Strategy for Inhibiting DNases. JACS AU 2024; 4:2262-2266. [PMID: 38938790 PMCID: PMC11200219 DOI: 10.1021/jacsau.4c00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 06/29/2024]
Abstract
This study highlights the novel potential of molecular aggregates as inhibitors of a disease-related protein. Enzyme inhibitors have been studied and developed as molecularly targeted drugs and have been applied for cancer, autoimmune diseases, and infections. In many cases, enzyme inhibitors that are used for therapeutic applications interact directly with enzymes in a molecule-to-molecule manner. We found that the aggregates of a small compound, Mn007, inhibited bovine pancreatic DNase I. Once Mn007 molecules formed aggregates, they exhibited inhibitory effects specific to DNases that require divalent metal ions. A DNase secreted by Streptococcus pyogenes causes streptococcal toxic shock syndrome (STSS). STSS is a severe infectious disease with a fatality rate exceeding 30% in patients, even in this century. S. pyogenes disrupts the human barrier system against microbial infections through the secreted DNase. Until now, the discovery/development of a DNase inhibitor has been challenging. Mn007 aggregates were found to inhibit the DNase secreted by S. pyogenes, which led to the successful suppression of S. pyogenes growth in human whole blood. To date, molecular aggregation has been outside the scope of drug discovery. The present study suggests that molecular aggregation is a vast area to be explored for drug discovery and development because aggregates of small-molecule compounds can inhibit disease-related enzymes.
Collapse
Affiliation(s)
- Kenta Morita
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
- Research
Center for Membrane and Film Technology, Kobe University, 1-1
Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Tomoko Moriwaki
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Shunsuke Habe
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Mariko Taniguchi-Ikeda
- Department
of Clinical Genetics, Fujita Health University
Hospital 1-98 Dengakugakubo, Kutsukake-chou, Toyoake, Aichi 470-1192, Japan
| | - Tadao Hasegawa
- Department
of Bacteriology, Graduate School of Medical
Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yusuke Minato
- Department
of Microbiology, School of Medicine, Fujita
Health University, 1-98
Dengakugakubo, Kutsukake-chou, Toyoake, Aichi 470-1192, Japan
| | - Takashi Aoi
- Division
of Stem Cell Medicine, Graduate School of Medicine, Kobe University, 7-5-1
Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tatsuo Maruyama
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
- Research
Center for Membrane and Film Technology, Kobe University, 1-1
Rokkodai, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
5
|
Boero E, Carducci M, Keeley AJ, Berlanda Scorza F, Iturriza-Gómara M, Moriel DG, Rossi O. A flow cytometry-based assay to determine the ability of anti-Streptococcus pyogenes antibodies to mediate monocytic phagocytosis in human sera. J Immunol Methods 2024; 528:113652. [PMID: 38458312 DOI: 10.1016/j.jim.2024.113652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Abstract
Streptococcus pyogenes, commonly referred to as Group A Streptococcus (Strep A), causes a spectrum of diseases, with the potential to progress into life-threatening illnesses and autoimmune complications. The escalating threat of antimicrobial resistance, stemming from the prevalent reliance on antibiotic therapies to manage Strep A infections, underscores the critical need for the development of disease control strategies centred around vaccination. Phagocytes play a critical role in controlling Strep A infections, and phagocytosis-replicating assays are essential for vaccine development. Traditionally, such assays have employed whole-blood killing or opsonophagocytic methods using HL-60 cells as neutrophil surrogates. However, assays mimicking Fcγ receptors- phagocytosis in clinical contexts are lacking. Therefore, here we introduce a flow cytometry-based method employing undifferentiated THP-1 cells as monocytic/macrophage model to swiftly evaluate the ability of human sera to induce phagocytosis of Strep A. We extensively characterize the assay's precision, linearity, and quantification limit, ensuring robustness. By testing human pooled serum, the assay proved to be suitable for the comparison of human sera's phagocytic capability against Strep A. This method offers a valuable complementary assay for clinical studies, addressing the gap in assessing FcγR-mediated phagocytosis. By facilitating efficient evaluation of Strep A -phagocyte interactions, it may contribute to elucidating the mechanisms required for the prevention of infections and inform the development of future vaccines and therapeutic advancements against Strep A infections.
Collapse
Affiliation(s)
- Elena Boero
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy.
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - Alexander J Keeley
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK; Vaccines and Immunity Theme, Medical Research Unit the Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Fajara, P. O. Box 273, the Gambia
| | | | - Miren Iturriza-Gómara
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - Danilo Gomes Moriel
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy
| |
Collapse
|
6
|
Vega LA, Sansón-Iglesias M, Mukherjee P, Buchan K, Morrison G, Hohlt AE, Flores AR. LiaR-dependent gene expression contributes to antimicrobial responses in group A Streptococcus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588141. [PMID: 38617309 PMCID: PMC11014544 DOI: 10.1101/2024.04.04.588141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The ability to sense and respond to host defenses is essential for pathogen survival. Some mechanisms involve two-component systems (TCS) that respond to host molecules, such as antimicrobial peptides (AMPs) and activate specific gene regulatory pathways to aid in survival. Alongside TCSs, bacteria coordinate cell division proteins, chaperones, cell wall sortases and secretory translocons at discrete locations within the cytoplasmic membrane, referred to as functional membrane microdomains (FMMs). In Group A Streptococcus (GAS), the FMM or "ExPortal" coordinates protein secretion, cell wall synthesis and sensing of AMP-mediated cell envelope stress via the LiaFSR three-component system. Previously we showed GAS exposure to a subset of AMPs (α-defensins) activates the LiaFSR system by disrupting LiaF and LiaS co-localization in the ExPortal, leading to increased LiaR phosphorylation, expression of the transcriptional regulator SpxA2, and altered GAS virulence gene expression. The mechanisms by which LiaFSR integrates cell envelope stress with responses to AMP activity and virulence are not fully elucidated. Here, we show the LiaFSR regulon is comprised of genes encoding SpxA2 and three membrane-associated proteins: a PspC domain-containing protein (PCP), the lipoteichoic acid-modifying protein LafB and the membrane protein insertase YidC2. Our data show phosphorylated LiaR induces transcription of these genes via a conserved operator, whose disruption attenuates GAS virulence and increases susceptibility to AMPs in a manner primarily dependent on differential expression of SpxA2. Our work expands understanding of the LiaFSR regulatory network in GAS and identifies targets for further investigation of mechanisms of cell envelope stress tolerance contributing to GAS pathogenesis.
Collapse
|
7
|
Opsonophagocytic Killing Assay to Measure Anti-Group A Streptococcus Antibody Functionality in Human Serum. Methods Mol Biol 2021. [PMID: 34784047 DOI: 10.1007/978-1-0716-1900-1_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
The opsonophagocytic killing assay (OPKA) is designed to measure the functionality of strain-specific antibodies and, therefore, assess protective immunity or the immunogenicity of Group A Streptococcus (GAS) (type A Streptococcus pyogenes) vaccines. Opsonization of GAS for phagocytosis is an important mechanism by which antibodies protect against disease in vivo. The Opsonophagocytic Index or Opsonic Index (OI) is the estimated dilution of antisera that kills 50% of the target bacteria. Here, we describe the protocol of the standardized GAS OPKA developed by Jones et al., 2018.
Collapse
|
8
|
Population Genomics of emm4 Group A Streptococcus Reveals Progressive Replacement with a Hypervirulent Clone in North America. mSystems 2021; 6:e0049521. [PMID: 34374563 PMCID: PMC8409732 DOI: 10.1128/msystems.00495-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Clonal replacement is a major driver for changes in bacterial disease epidemiology. Recently, it has been proposed that episodic emergence of novel, hypervirulent clones of group A Streptococcus (GAS) results from acquisition of a 36-kb DNA region leading to increased expression of the cytotoxins Nga (NADase) and SLO (streptolysin O). We previously described a gene fusion event involving the gene encoding the GAS M protein (emm) and an adjacent M-like protein (enn) in the emm4 GAS population, a GAS emm type that lacks the hyaluronic acid capsule. Using whole-genome sequencing of a temporally and geographically diverse set of 1,126 isolates, we discovered that the North American emm4 GAS population has undergone clonal replacement with emergent GAS strains completely replacing historical isolates by 2017. Emergent emm4 GAS strains contained a handful of small genetic variations, including the emm-enn gene fusion, and showed a marked in vitro growth defect compared to historical strains. In contrast to other previously described GAS clonal replacement events, emergent emm4 GAS strains were not defined by acquisition of exogenous DNA and had no significant increase in transcript levels of nga and slo toxin genes via RNA sequencing and quantitative real-time PCR analysis relative to historic strains. Despite the in vitro growth differences, emergent emm4 GAS strains were hypervirulent in mice and ex vivo growth in human blood compared to historical strains. Thus, these data detail the emergence and dissemination of a hypervirulent acapsular GAS clone defined by small, endogenous genetic variation, thereby defining a novel model for GAS strain replacement. IMPORTANCE Severe invasive infections caused by group A Streptococcus (GAS) result in substantial morbidity and mortality in children and adults worldwide. Previously, GAS clonal strain replacement has been attributed to acquisition of exogenous DNA leading to novel virulence gene acquisition or increased virulence gene expression. Our study of type emm4 GAS identified emergence of a hypervirulent GAS clade defined by variation in endogenous DNA content and lacking augmented toxin gene expression relative to replaced strains. These findings expand our understanding of the molecular mechanisms underlying bacterial clonal emergence.
Collapse
|
9
|
The LiaFSR transcriptome reveals an interconnected regulatory network in group A Streptococcus. Infect Immun 2021; 89:e0021521. [PMID: 34370508 DOI: 10.1128/iai.00215-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanisms by which bacteria sense the host environment and alter gene expression are poorly understood. LiaFSR is a gene regulatory system unique to Gram-positive bacteria including group A Streptococcus (GAS) and responds to cell envelope stress. We previously showed that LiaF acts as an inhibitor to LiaFSR activation in GAS. To better understand gene regulation associated with LiaFSR activation, we performed RNA-sequencing on isogenic deletion mutants fixed in a LiaFSR "always on" (ΔliaF) or "always off" (ΔliaR) state. Transcriptome analyses of ΔliaF and ΔliaR in GAS showed near perfect inverse correlation including the gene encoding the global transcriptional regulator SpxA2. In addition, mutant transcriptomes included genes encoding multiple virulence factors and showed substantial overlap with the CovRS regulon. Chromatin immunoprecipitation quantitative PCR demonstrated direct spxA2 gene regulation following activation of the response regulator, LiaR. High SpxA2 levels as a result of LiaFSR activation were directly correlated with increased CovR-regulated virulence gene transcription. Further, consistent with known virulence gene repression by phosphorylated CovR, elevated SpxA2 levels were inversely correlated with CovR phosphorylation. Despite increased transcription of several virulence factors, ΔliaF (high SpxA2) exhibited a paradoxical virulence phenotype in both in vivo mouse and ex vivo human blood models of disease. Likewise, despite decreased virulence factor transcription in ΔliaR (low SpxA2), increased virulence was observed in an in vivo mouse model of disease - a phenotype attributable, in part, to known SpxA2-associated speB transcription. Our findings provide evidence of a critical role of LiaFSR in sensing the host environment and suggest a potential mechanism for gene regulatory system crosstalk shared by many Gram-positive pathogens.
Collapse
|
10
|
Bläckberg A, de Neergaard T, Frick IM, Nordenfelt P, Lood R, Rasmussen M. Lack of Opsonic Antibody Responses to Invasive Infections With Streptococcus dysgalactiae. Front Microbiol 2021; 12:635591. [PMID: 33986732 PMCID: PMC8111088 DOI: 10.3389/fmicb.2021.635591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/31/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction Streptococcus dysgalactiae can cause severe recurrent infections. This study aimed to investigate antibody responses following S. dysgalactiae bacteraemia and possible development of protective immunity. Materials and Methods Patients with S. dysgalactiae bacteraemia in the county of Skåne between 2017 and 2018 were prospectively included. Acute and convalescent sera were obtained. All isolates were emm typed and enzyme-linked immunosorbent assay (ELISA) was utilised to analyse specific antibody responses to bacteria and antigens. Bactericidal- and phagocytosis assays were applied to further establish antibody function. Results Sixteen patients with S. dysgalactiae bacteraemia were included of whom one had recurrent episodes of bacteraemia. Using ELISA with S. dysgalactiae isolates and mutants, development of IgG antibodies was demonstrated in few patients. Type-specific antibodies were demonstrated in one patient when recombinant M proteins as antigens, were applied. The type-specific serum mediated a small increase in phagocytosis but did not facilitate increased killing of the S. dysgalactiae isolate, carrying that M protein, in blood or by phagocytic cells. Conclusion S. dysgalactiae bacteraemia sometimes results in increased levels of antibodies to the infecting pathogen. We did not find evidence that these antibodies are effectively opsonising. Apparent failure to produce opsonising antibodies might partially explain why S. dysgalactiae can cause recurrent invasive infections in the same host.
Collapse
Affiliation(s)
- Anna Bläckberg
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Skåne University Hospital, Department of Infectious Diseases, Lund, Sweden
| | - Therese de Neergaard
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Inga-Maria Frick
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Rolf Lood
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Magnus Rasmussen
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Skåne University Hospital, Department of Infectious Diseases, Lund, Sweden
| |
Collapse
|
11
|
Wierzbicki IH, Campeau A, Dehaini D, Holay M, Wei X, Greene T, Ying M, Sands JS, Lamsa A, Zuniga E, Pogliano K, Fang RH, LaRock CN, Zhang L, Gonzalez DJ. Group A Streptococcal S Protein Utilizes Red Blood Cells as Immune Camouflage and Is a Critical Determinant for Immune Evasion. Cell Rep 2020; 29:2979-2989.e15. [PMID: 31801066 PMCID: PMC6951797 DOI: 10.1016/j.celrep.2019.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 09/09/2019] [Accepted: 10/30/2019] [Indexed: 01/17/2023] Open
Abstract
Group A Streptococcus (GAS) is a human-specific pathogen that evades the host immune response through the elaboration of multiple virulence factors. Although many of these factors have been studied, numerous proteins encoded by the GAS genome are of unknown function. Herein, we characterize a biomimetic red blood cell (RBC)-captured protein of unknown function—annotated subsequently as S protein—in GAS pathophysiology. S protein maintains the hydrophobic properties of GAS, and its absence reduces survival in human blood. S protein facilitates GAS coating with lysed RBCs to promote molecular mimicry, which increases virulence in vitro and in vivo. Proteomic profiling reveals that the removal of S protein from GAS alters cellular and extracellular protein landscapes and is accompanied by a decrease in the abundance of several key GAS virulence determinants. In vivo, the absence of S protein results in a striking attenuation of virulence and promotes a robust immune response and immunological memory. Wierzbicki et al. show that S protein is a major group A Streptococcus (GAS) virulence factor that facilitates bacterial coating with lysed red blood cells to promote molecular mimicry, which increases virulence in vitro and in vivo. Removal of S protein reduces the abundance of multiple virulence factors and attenuates virulence.
Collapse
Affiliation(s)
- Igor H Wierzbicki
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anaamika Campeau
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Diana Dehaini
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maya Holay
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiaoli Wei
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Trever Greene
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Man Ying
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jenna S Sands
- Department of Microbiology and Immunology, Division of Infectious Diseases, and Antimicrobial Resistance Center, Emory University, Atlanta, GA 30322, USA
| | - Anne Lamsa
- Department of Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elina Zuniga
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kit Pogliano
- Department of Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christopher N LaRock
- Department of Microbiology and Immunology, Division of Infectious Diseases, and Antimicrobial Resistance Center, Emory University, Atlanta, GA 30322, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - David J Gonzalez
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
12
|
Phosphotransferase System Uptake and Metabolism of the β-Glucoside Salicin Impact Group A Streptococcal Bloodstream Survival and Soft Tissue Infection. Infect Immun 2020; 88:IAI.00346-20. [PMID: 32719156 DOI: 10.1128/iai.00346-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Streptococcus pyogenes (group A Streptococcus [GAS]), a major human-specific pathogen, relies on efficient nutrient acquisition for successful infection within its host. The phosphotransferase system (PTS) couples the import of carbohydrates with their phosphorylation prior to metabolism and has been linked to GAS pathogenesis. In a screen of an insertional mutant library of all 14 annotated PTS permease (EIIC) genes in MGAS5005, the annotated β-glucoside PTS transporter (bglP) was found to be crucial for GAS growth and survival in human blood and was validated in another M1T1 GAS strain, 5448. In 5448, bglP was shown to be in an operon with a putative phospho-β-glucosidase (bglB) downstream and a predicted antiterminator (licT) upstream. Using defined nonpolar mutants of the β-glucoside permease (bglP) and β-glucosidase enzyme (bglB) in 5448, we showed that bglB, not bglP, was important for growth in blood. Furthermore, transcription of the licT-blgPB operon was found to be repressed by glucose and induced by the β-glucoside salicin as the sole carbon source. Investigation of the individual bglP and bglB mutants determined that they influence in vitro growth in the β-glucoside salicin; however, only bglP was necessary for growth in other non-β-glucoside PTS sugars, such as fructose and mannose. Additionally, loss of BglP and BglB suggests that they are important for the regulation of virulence-related genes that control biofilm formation, streptolysin S (SLS)-mediated hemolysis, and localized ulcerative lesion progression during subcutaneous infections in mice. Thus, our results indicate that the β-glucoside PTS transports salicin and its metabolism can differentially influence GAS pathophysiology during soft tissue infection.
Collapse
|
13
|
Eraso JM, Kachroo P, Olsen RJ, Beres SB, Zhu L, Badu T, Shannon S, Cantu CC, Saavedra MO, Kubiak SL, Porter AR, DeLeo FR, Musser JM. Genetic heterogeneity of the Spy1336/R28-Spy1337 virulence axis in Streptococcus pyogenes and effect on gene transcript levels and pathogenesis. PLoS One 2020; 15:e0229064. [PMID: 32214338 PMCID: PMC7098570 DOI: 10.1371/journal.pone.0229064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/28/2020] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pyogenes is a strict human pathogen responsible for more than 700 million infections annually worldwide. Strains of serotype M28 S. pyogenes are typically among the five more abundant types causing invasive infections and pharyngitis in adults and children. Type M28 strains also have an unusual propensity to cause puerperal sepsis and neonatal disease. We recently discovered that a one-nucleotide indel in an intergenic homopolymeric tract located between genes Spy1336/R28 and Spy1337 altered virulence in a mouse model of infection. In the present study, we analyzed size variation in this homopolymeric tract and determined the extent of heterogeneity in the number of tandemly-repeated 79-amino acid domains in the coding region of Spy1336/R28 in large samples of strains recovered from humans with invasive infections. Both repeat sequence elements are highly polymorphic in natural populations of M28 strains. Variation in the homopolymeric tract results in (i) changes in transcript levels of Spy1336/R28 and Spy1337 in vitro, (ii) differences in virulence in a mouse model of necrotizing myositis, and (iii) global transcriptome changes as shown by RNAseq analysis of isogenic mutant strains. Variation in the number of tandem repeats in the coding sequence of Spy1336/R28 is responsible for size variation of R28 protein in natural populations. Isogenic mutant strains in which genes encoding R28 or transcriptional regulator Spy1337 are inactivated are significantly less virulent in a nonhuman primate model of necrotizing myositis. Our findings provide impetus for additional studies addressing the role of R28 and Spy1337 variation in pathogen-host interactions.
Collapse
Affiliation(s)
- Jesus M. Eraso
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Priyanka Kachroo
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Randall J. Olsen
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
- Departments of Pathology and Laboratory Medicine and Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Stephen B. Beres
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Luchang Zhu
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Traci Badu
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Sydney Shannon
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Concepcion C. Cantu
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Matthew Ojeda Saavedra
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Samantha L. Kubiak
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
| | - Adeline R. Porter
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Frank R. DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - James M. Musser
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Houston Methodist Hospital, Houston, Texas, United States of America
- Departments of Pathology and Laboratory Medicine and Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
14
|
Strain-Dependent Effect of Capsule on Transmission and Persistence in an Infant Mouse Model of Group A Streptococcus Infection. Infect Immun 2020; 88:IAI.00709-19. [PMID: 32014891 DOI: 10.1128/iai.00709-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus [GAS]) is a human pathogen responsible for a wide range of diseases. Asymptomatic carriage of GAS in the human pharynx is commonplace and a potential reservoir for GAS transmission. Early studies showed that GAS transmission correlated with high bacterial burdens during the acute symptomatic phase of the disease. Human studies and the nonhuman primate model are generally impractical for investigation of the bacterial mechanisms contributing to GAS transmission and persistence. To address this gap, we adapted an infant mouse model of pneumococcal colonization and transmission to investigate factors that influence GAS transmission and persistence. The model recapitulated the direct correlation between GAS burden and transmission during the acute phase of infection observed in humans and nonhuman primates. Furthermore, our results indicate that the ratio of colonized to uncolonized hosts influences the rates of GAS transmission and persistence. We used the model to test the hypothesis that capsule production influences GAS transmission and persistence in a strain-dependent manner. We detected significant differences in rates of transmission and persistence between capsule-positive (emm3) and capsule-negative (emm87) GAS strains. Capsule was associated with higher levels of GAS shedding, independent of the strain background. In contrast to the capsule-positive emm3 strain, restoring capsule production in emm87 GAS did not increase transmissibility, and the absence of capsule enhanced persistence only in the capsule-negative (emm87) strain background. These data suggest that strain background (capsule positive versus capsule negative) influences the effect of capsule in GAS transmission and persistence and that as-yet-undefined factors are required for the transmission of capsule-negative emm types.
Collapse
|
15
|
McGregor R, Jones S, Jeremy RM, Goldblatt D, Moreland NJ. An Opsonophagocytic Killing Assay for the Evaluation of Group A Streptococcus Vaccine Antisera. Methods Mol Biol 2020; 2136:323-335. [PMID: 32430834 DOI: 10.1007/978-1-0716-0467-0_26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Group A Streptococcus (GAS) is a major cause of global mortality, yet there are no licensed GAS vaccines. Vaccine progress has been hampered, in part, by a lack of standardized assays able to quantify antibody function in test antisera. The most widely used assay was developed over 50 years ago by Rebecca Lancefield and relies on human whole blood as a source of complement and neutrophils. Recently, an opsonophagocytic killing (OPK) assay has been developed for GAS by adapting the OPK methods utilized in Streptococcus pneumoniae vaccine testing. This assay uses dimethylformamide (DMF)-differentiated human promyelocytic leukemia cells (HL-60 cells) as a source of neutrophils and baby rabbit complement, thus removing the major sources of variation in the Lancefield assays. This protocol outlines methods for performing a GAS OPK assay including titering test sera to generate an opsonic index. This in vitro assay could aid in selecting vaccine candidates by demonstrating whether candidate-induced antibodies lead to complement deposition and opsonophagocytic killing.
Collapse
Affiliation(s)
- Reuben McGregor
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.
| | - Scott Jones
- Immunobiology, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Raynes M Jeremy
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - David Goldblatt
- Immunobiology, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nicole J Moreland
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Lancefield Whole Blood Killing Assay to Evaluate Vaccine Efficacy. Methods Mol Biol 2020; 2136:317-322. [PMID: 32430833 DOI: 10.1007/978-1-0716-0467-0_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
While the Lancefield whole blood killing assay is named after the renowned streptococcal researcher Rebecca Lancefield, the protocol was first described by Todd in 1927 (Br J Exp Pathol 8:1-5, 1927). Initially, the assay was used to identify novel Group A Streptococcal (GAS) serotypes through the supplementation of non-immune human blood (often from infants) with type-specific antisera prepared in rabbits (Lancefield, J Exp Med 106:525-544, 1957; Maxted, Br J Exp Pathol 37:415-422, 1956) and to demonstrate the impressive longevity of type-specific immunity in patients following invasive GAS infection (Lancefield, J Exp Med 110:271-292, 1959). The modern assay is routinely used to screen defined GAS mutants (Wessels, Bronze, Proc Natl Acad Sci U S A 91:12238-12242, 1994; Zinkernagel et al., Cell Host Microbe 4:170-178, 2008) or transposon libraries (Le Breton et al., Infect Immun 81:862-875, 2013) for enhanced susceptibility to opsonophagocytic killing or to screen vaccine antisera (Salehi et al., mSphere 3:e00617-e00618, 2018) or other serological preparations (Reglinski et al., Sci Rep 5:15825, 2015) for anti-streptococcal activity.
Collapse
|
17
|
Pastural É, McNeil SA, MacKinnon-Cameron D, Ye L, Langley JM, Stewart R, Martin LH, Hurley GJ, Salehi S, Penfound TA, Halperin S, Dale JB. Safety and immunogenicity of a 30-valent M protein-based group a streptococcal vaccine in healthy adult volunteers: A randomized, controlled phase I study. Vaccine 2019; 38:1384-1392. [PMID: 31843270 DOI: 10.1016/j.vaccine.2019.12.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Streptococcus pyogenes (group A Streptococcus, Strep A) is a widespread pathogen that continues to pose a significant threat to human health. The development of a Strep A vaccine remains an unmet global health need. One of the major vaccine strategies is the use of M protein, which is a primary virulence determinant and protective antigen. Multivalent recombinant M protein vaccines are being developed with N-terminal M peptides that contain opsonic epitopes but do not contain human tissue cross-reactive epitopes. METHODS We completed a Phase I trial of a recombinant 30-valent M protein-based Strep A vaccine (Strep A vaccine, StreptAnova™) comprised of four recombinant proteins containing N-terminal peptides from 30 M proteins of common pharyngitis and invasive and/or rheumatogenic serotypes, adjuvanted with aluminum hydroxide. The trial was observer-blinded and randomized in a 2:1 ratio for intramuscular administration of Strep A vaccine or an alum-based comparator in healthy adult volunteers, at 0, 30 and 180 days. Primary outcome measures were assessments of safety, including assays for antibodies that cross-reacted with host tissues, and immunogenicity assessed by ELISA with the individual vaccine peptides and by opsonophagocytic killing (OPK) assays in human blood. RESULTS Twenty-three Strep A-vaccinated participants and 13 controls completed the study. The Strep A vaccine was well-tolerated and there was no clinical evidence of autoimmunity and no laboratory evidence of tissue cross-reactive antibodies. The vaccine was immunogenic and elicited significant increases in geometric mean antibody levels to 24 of the 30 component M antigens by ELISA. Vaccine-induced OPK activity was observed against selected M types of Strep A in vaccinated participants that seroconverted to specific M peptides. CONCLUSION The Strep A vaccine was well tolerated and immunogenic in healthy adults, providing strong support for further clinical development. [ClinicalTrials.gov NCT02564237].
Collapse
Affiliation(s)
- Élodie Pastural
- Pan-Provincial Vaccine Enterprise Inc. (PREVENT), Saskatoon, Saskatchewan, Canada
| | - Shelly A McNeil
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada; Division of Infectious Diseases, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Donna MacKinnon-Cameron
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Lingyun Ye
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Joanne M Langley
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada; Division of Infectious Diseases, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Robert Stewart
- Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Luis H Martin
- Pan-Provincial Vaccine Enterprise Inc. (PREVENT), Saskatoon, Saskatchewan, Canada
| | - Gregory J Hurley
- Division of Infectious Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Salehi
- Division of Infectious Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thomas A Penfound
- Division of Infectious Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Scott Halperin
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada; Division of Infectious Diseases, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - James B Dale
- Division of Infectious Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
18
|
The scfCDE Operon Encodes a Predicted ABC Importer Required for Fitness and Virulence during Group A Streptococcus Invasive Infection. Infect Immun 2019; 87:IAI.00613-19. [PMID: 31591169 DOI: 10.1128/iai.00613-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/27/2019] [Indexed: 02/08/2023] Open
Abstract
As a strict human pathogen, Streptococcus pyogenes (group A Streptococcus, or GAS) causes a wide range of infections, from superficial to life-threatening diseases, upon dissemination. Thus, it is necessary to gain a better understanding of how GAS successfully overcomes host-mediated challenges and infects various host niches. We previously identified subcutaneous fitness (scf) genes in the clinically relevant wild-type (WT) GAS M1T1 5448 strain that are critical for fitness during murine soft-tissue infection at both 24 h and 48 h postinfection. The uncharacterized locus scfCDE was transcribed as an operon and is predicted to encode an ABC importer for nutrient uptake (e.g., amino acids). Individual scfCDE deletion mutants grew comparably to WT 5448 in rich medium but exhibited reduced fitness during competitive growth in murine soft tissue and in nutrient-limiting chemically defined medium (CDM). A deletion of the permease gene scfD resulted in a monoculture growth defect in CDM that could be rescued by addition of excess peptides, suggesting a role as an amino acid importer. Interestingly, the ΔscfC substrate-binding and ΔscfD permease mutants, but not the ΔscfE ATPase mutant, were highly attenuated in murine soft tissue. Moreover, all three genes were required for GAS survival in human blood, indicating their impact is not limited to superficial infections. As such, scfCDE plays an integral role in enhancing GAS adaptation during localized infection as well as dissemination to deeper host environments. Since scfCDE is conserved throughout Firmicutes, this work may contribute to the development of therapeutic strategies against GAS and other Gram-positive pathogens.
Collapse
|
19
|
Naegeli A, Bratanis E, Karlsson C, Shannon O, Kalluru R, Linder A, Malmström J, Collin M. Streptococcus pyogenes evades adaptive immunity through specific IgG glycan hydrolysis. J Exp Med 2019; 216:1615-1629. [PMID: 31092533 PMCID: PMC6605743 DOI: 10.1084/jem.20190293] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 12/19/2022] Open
Abstract
EndoS from Streptococcus pyogenes hydrolyzes the functionally important glycan on the Fc portion of IgG during infections in humans. In mice with IgG mediated immunity against the M1 protein on the bacteria, EndoS is a virulence factor. Streptococcus pyogenes (Group A streptococcus; GAS) is a human pathogen causing diseases from uncomplicated tonsillitis to life-threatening invasive infections. GAS secretes EndoS, an endoglycosidase that specifically cleaves the conserved N-glycan on IgG antibodies. In vitro, removal of this glycan impairs IgG effector functions, but its relevance to GAS infection in vivo is unclear. Using targeted mass spectrometry, we characterized the effects of EndoS on host IgG glycosylation during the course of infections in humans. Substantial IgG glycan hydrolysis occurred at the site of infection and systemically in the severe cases. We demonstrated decreased resistance to phagocytic killing of GAS lacking EndoS in vitro and decreased virulence in a mouse model of invasive infection. This is the first described example of specific bacterial IgG glycan hydrolysis during infection and thereby verifies the hypothesis that EndoS modifies antibodies in vivo. This mechanisms of immune evasion could have implications for treatment of severe GAS infections and for future efforts at vaccine development.
Collapse
Affiliation(s)
- Andreas Naegeli
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Eleni Bratanis
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Christofer Karlsson
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Oonagh Shannon
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Raja Kalluru
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Adam Linder
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Johan Malmström
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Mattias Collin
- Faculty of Medicine, Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| |
Collapse
|
20
|
Hirose Y, Yamaguchi M, Goto K, Sumitomo T, Nakata M, Kawabata S. Competence-induced protein Ccs4 facilitates pneumococcal invasion into brain tissue and virulence in meningitis. Virulence 2019; 9:1576-1587. [PMID: 30251911 PMCID: PMC6177246 DOI: 10.1080/21505594.2018.1526530] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Streptococcus pneumoniae is a major pathogen that causes pneumonia, sepsis, and meningitis. The candidate combox site 4 (ccs4) gene has been reported to be a pneumococcal competence-induced gene. Such genes are involved in development of S. pneumoniae competence and virulence, though the functions of ccs4 remain unknown. In the present study, the role of Ccs4 in the pathogenesis of pneumococcal meningitis was examined. We initially constructed a ccs4 deletion mutant and complement strains, then examined their association with and invasion into human brain microvascular endothelial cells. Wild-type and Ccs4-complemented strains exhibited significantly higher rates of association and invasion as compared to the ccs4 mutant strain. Deletion of ccs4 did not change bacterial growth activity or expression of NanA and CbpA, known brain endothelial pneumococcal adhesins. Next, mice were infected either intravenously or intranasally with pneumococcal strains. In the intranasal infection model, survival rates were comparable between wild-type strain-infected and ccs4 mutant strain-infected mice, while the ccs4 mutant strain exhibited a lower level of virulence in the intravenous infection model. In addition, at 24 hours after intravenous infection, the bacterial burden in blood was comparable between the wild-type and ccs4 mutant strain-infected mice, whereas the wild-type strain-infected mice showed a significantly higher bacterial burden in the brain. These results suggest that Ccs4 contributes to pneumococcal invasion of host brain tissues and functions as a virulence factor.
Collapse
Affiliation(s)
- Yujiro Hirose
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Masaya Yamaguchi
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Kana Goto
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Tomoko Sumitomo
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Masanobu Nakata
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Shigetada Kawabata
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| |
Collapse
|
21
|
Wessels MR. Capsular Polysaccharide of Group A Streptococcus. Microbiol Spectr 2019; 7:10.1128/microbiolspec.GPP3-0050-2018. [PMID: 30632480 PMCID: PMC6342470 DOI: 10.1128/microbiolspec.gpp3-0050-2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Indexed: 01/02/2023] Open
Abstract
Most clinical isolates of Streptococcus pyogenes elaborate a capsular polysaccharide, which is composed of hyaluronic acid, a high-molecular-mass polymer of alternating residues of N-acetyl glucosamine and glucuronic acid. Certain strains, particularly those of the M18 serotype, produce abundant amounts of capsule, resulting in formation of large, wet-appearing, translucent or "mucoid" colonies on solid media, whereas strains of M-types 4 and 22 produce none. Studies of acapsular mutant strains have provided evidence that the capsule enhances virulence in animal models of infection, an effect attributable, at least in part, to resistance to complement-mediated opsonophagocytic killing by leukocytes. The presence of the hyaluronic acid capsule may mask adhesins on the bacterial cell wall. However, the capsule itself can mediate bacterial attachment to host cells by binding to the hyaluronic-acid binding protein, CD44. Furthermore, binding of the S. pyogenes capsule to CD44 on host epithelial cells can trigger signaling events that disrupt cell-cell junctions and facilitate bacterial invasion into deep tissues. This article summarizes the biochemistry, genetics, regulation, and role in pathogenesis of this important virulence determinant.
Collapse
Affiliation(s)
- Michael R Wessels
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Development of an Opsonophagocytic Killing Assay Using HL-60 Cells for Detection of Functional Antibodies against Streptococcus pyogenes. mSphere 2018; 3:3/6/e00617-18. [PMID: 30567901 PMCID: PMC6300688 DOI: 10.1128/msphere.00617-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The clinical development of group A streptococcal (GAS) vaccines will require the implementation of a standardized, high-throughput assay to measure the activity of functional opsonic antibodies in vaccine recipients. In the present study, we adapted and modified the HL-60-based protocol that was developed for the detection of opsonic antibodies against Streptococcus pneumoniae for use with multiple M types of GAS. Modifications of the assay conditions permitted the evaluation of 21 different M types of GAS in the assay. The specificity of the antibody-mediated opsonization was demonstrated by inhibition with homologous, but not heterologous, M proteins. Maximum rates of opsonophagocytic killing (OPK) of 14 different M types promoted by rabbit antiserum against the 30-valent M protein-based vaccine were comparable in whole-blood and HL-60 assays. Data are also presented showing OPK serum titers (opsonic index) of naturally acquired human antibodies present in IVIG [intravenous immune globulin (human)]. Results of the HL-60 assay performed on different days using 21 different M types of GAS and IVIG as the antibody source were significantly concordant. This report indicates that the OPK assay conditions may be optimized for the measurement of opsonic antibodies against a number of epidemiologically important M types of GAS and, once standardized, should facilitate the clinical development of effective vaccines to prevent these infections.IMPORTANCE Measuring functional opsonic antibodies against group A streptococci is an important component of the clinical development path for effective vaccines. Prior studies have used an assay developed over 60 years ago that relied on whole human blood as the source of phagocytes and complement, both of which are critical components of antibody-mediated killing assays. In this study, we adapted an assay that uses the HL-60 human promyelocytic leukemia cell line as phagocytic cells and baby rabbit serum as a source of complement for detection of opsonic antibodies against group A streptococci. On the basis of some of the known biological characteristics of the bacteria, we modified the assay conditions to support the evaluation of 21 epidemiologically important M types and demonstrated the utility and reproducibility of the assay for measurement of functional opsonic antibody levels.
Collapse
|
23
|
Zhu W, Wu C, Kang C, Cai C, Wang Y, Li J, Zhang Q, Sun X, Jin M. Evaluation of the protective efficacy of four newly identified surface proteins of Erysipelothrix rhusiopathiae. Vaccine 2018; 36:8079-8083. [PMID: 30446176 DOI: 10.1016/j.vaccine.2018.10.071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/15/2018] [Accepted: 10/21/2018] [Indexed: 10/27/2022]
Abstract
Erysipelothrix rhusiopathiae is the causative agent of animal erysipelas and human erysipeloid. Bacterial surface proteins are promising vaccine candidates. We recently identified 3 E. rhusiopathiae surface proteins (GAPDH, HP0728, and HP1472) and characterized their roles as virulence factors. However, their efficacy as protective antigens is still unknown. The N-terminal region of a previously identified surface protein, CbpB (CbpB-N), is speculated to be a protective antigen, but this needs to be verified. The aim of this study was to evaluate the protective efficacy of GAPDH, HP0728, HP1472, and CbpB-N. Immunization with recombinant GAPDH provided complete protection in a mouse model, recombinant CbpB-N provided partial protection, while recombinant HP0728 and HP1472 provided no protection. Recombinant GAPDH also provided good protection in a pig model. GAPDH antiserum exhibited significant blood bactericidal activity against E. rhusiopathiae. In conclusion, GAPDH and CbpB-N were found to be protective antigens of E. rhusiopathiae, and GAPDH is a promising vaccine candidate.
Collapse
Affiliation(s)
- Weifeng Zhu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chao Wu
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, China; Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Chao Kang
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengzhi Cai
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ya Wang
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jingtao Li
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiang Zhang
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Life Sciences & Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaomei Sun
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, China
| | - Meilin Jin
- Animal Infectious Disease Unit, National State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, China; Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.
| |
Collapse
|
24
|
Jones S, Moreland NJ, Zancolli M, Raynes J, Loh JMS, Smeesters PR, Sriskandan S, Carapetis JR, Fraser JD, Goldblatt D. Development of an opsonophagocytic killing assay for group a streptococcus. Vaccine 2018; 36:3756-3763. [PMID: 29776751 DOI: 10.1016/j.vaccine.2018.05.056] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/17/2018] [Accepted: 05/11/2018] [Indexed: 10/14/2022]
Abstract
Group A Streptococcus (GAS) or Streptococcus pyogenes is responsible for an estimated 500,000 deaths worldwide each year. Protection against GAS infection is thought to be mediated by phagocytosis, enhanced by bacteria-specific antibody. There are no licenced GAS vaccines, despite many promising candidates in preclinical and early stage clinical development, the most advanced of which are based on the GAS M-protein. Vaccine progress has been hindered, in part, by the lack of a standardised functional assay suitable for vaccine evaluation. Current assays, developed over 50 years ago, rely on non-immune human whole blood as a source of neutrophils and complement. Variations in complement and neutrophil activity between donors result in variable data that is difficult to interpret. We have developed an opsonophagocytic killing assay (OPKA) for GAS that utilises dimethylformamide (DMF)-differentiated human promyelocytic leukemia cells (HL-60) as a source of neutrophils and baby rabbit complement, thus removing the major sources of variation in current assays. We have standardised the OPKA for several clinically relevant GAS strain types (emm1, emm6 and emm12) and have shown antibody-specific killing for each emm-type using M-protein specific rabbit antisera. Specificity was demonstrated by pre-incubation of the antisera with homologous M-protein antigens that blocked antibody-specific killing. Additional qualifications of the GAS OPKA, including the assessment of the accuracy, precision, linearity and the lower limit of quantification, were also performed. This GAS OPKA assay has the potential to provide a robust and reproducible platform to accelerate GAS vaccine development.
Collapse
Affiliation(s)
- Scott Jones
- Immunobiology, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, United Kingdom.
| | - Nicole J Moreland
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Marta Zancolli
- Immunobiology, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | - Jeremy Raynes
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Pierre R Smeesters
- Molecular Bacteriology Laboratory, Universite ́ Libre de Bruxelles and Academic Children Hospital, Brussels, Belgium; Murdoch Children's Research Institute and University of Melbourne, Melbourne, Australia
| | - Shiranee Sriskandan
- Faculty of Medicine, Imperial College London, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - Jonathan R Carapetis
- Telethon Kids Institute, University of Western Australia and Perth Children's Hospital, Perth, Australia
| | - John D Fraser
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - David Goldblatt
- Immunobiology, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, United Kingdom
| |
Collapse
|
25
|
Frost HR, Laho D, Sanderson-Smith ML, Licciardi P, Donath S, Curtis N, Kado J, Dale JB, Steer AC, Smeesters PR. Immune Cross-Opsonization Within emm Clusters Following Group A Streptococcus Skin Infection: Broadening the Scope of Type-Specific Immunity. Clin Infect Dis 2018; 65:1523-1531. [PMID: 29020160 PMCID: PMC7263703 DOI: 10.1093/cid/cix599] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
Background Group AStreptococcus (GAS) skin infections are particularly prevalent in developing nations. The GAS M protein, by which strains are differentiated into >220 differentemm types, is immunogenic and elicits protective antibodies. A major obstacle for vaccine development has been the traditional understanding that immunity following infection is restricted to a singleemm type. However, recent evidence has led to the hypothesis of immune cross-reactivity betweenemm types. Methods We investigated the human serological response to GAS impetigo in Fijian schoolchildren, focusing on 3 majoremm clusters (E4, E6, and D4). Pre- and postinfection sera were assayed by enzyme-linked immunosorbent assay with N-terminal M peptides and bactericidal assays using the infecting-type strain,emm cluster–related strains, and nonrelated strains. Results Twenty of the 53 paired sera demonstrated a ≥4-fold increase in antibody titer against the infecting type. When tested against all cluster-related M peptides, we found that 9 of 17 (53%) paired sera had a ≥4-fold increase in antibody titer to cluster-related strains as well. When grouped by cluster, the mean change to cluster-relatedemm types in E4 and E6 was >4-fold (5.9-fold and 19.5-fold, respectively) but for D4 was 3.8-fold. The 17 paired sera were tested in bactericidal assays against selected cluster-related and nonrelated strains. While the responses were highly variable, numerous instances of cross-reactive killing were observed. Conclusions These data demonstrate that M type–specific and cross-reactive immune responses occur following skin infection. The cross-reactive immune responses frequently align withemm clusters, raising new opportunities to design multivalent vaccines with broad coverage.
Collapse
Affiliation(s)
- Hannah R Frost
- Group A Streptococcus Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,Molecular Bacteriology Laboratory
| | - Delphine Laho
- Group A Streptococcus Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,Department of Pediatrics, Academic Children Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Martina L Sanderson-Smith
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong
| | - Paul Licciardi
- Pneumococcal Research Group, Murdoch Childrens Research Institute, Melbourne.,Department of Paediatrics, University of Melbourne, Royal Children's Hospital Melbourne, Parkville, Australia
| | - Susan Donath
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital Melbourne, Parkville, Australia
| | - Nigel Curtis
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital Melbourne, Parkville, Australia
| | - Joseph Kado
- Department of Paediatrics, Colonial War Memorial Hospital.,College of Medicine, Nursing and Health Sciences, Fiji National University.,Fiji Rheumatic Heart Disease Control Program, Suva, Fiji
| | - James B Dale
- Medicine.,Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center.,Department of Veterans Affairs Medical Center, Memphis, Tennessee
| | - Andrew C Steer
- Group A Streptococcus Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Royal Children's Hospital Melbourne, Parkville, Australia.,Centre for International Child Health, University of Melbourne, Australia
| | - Pierre R Smeesters
- Group A Streptococcus Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,Molecular Bacteriology Laboratory.,Department of Pediatrics, Academic Children Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium.,Centre for International Child Health, University of Melbourne, Australia
| |
Collapse
|
26
|
Sundar GS, Islam E, Braza RD, Silver AB, Le Breton Y, McIver KS. Route of Glucose Uptake in the Group a Streptococcus Impacts SLS-Mediated Hemolysis and Survival in Human Blood. Front Cell Infect Microbiol 2018; 8:71. [PMID: 29594067 PMCID: PMC5861209 DOI: 10.3389/fcimb.2018.00071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
The transport and metabolism of glucose has been shown to have far reaching consequences in the transcriptional profile of many bacteria. As glucose is most often the preferred carbon source for bacteria, its presence in the environment leads to the repression of many alternate carbohydrate pathways, a condition known as carbon catabolite repression (CCR). Additionally, the expression of many virulence factors is also dependent on the presence of glucose. Despite its importance, little is known about the transport routes of glucose in the human pathogen Streptococcus pyogenes. Considering that Streptococcus pyogenes is an important human pathogen responsible for over 500,000 deaths every year, we characterized the routes of glucose transport in an effort to understand its importance in GAS pathogenesis. Using a deletion of glucokinase (ΔnagC) to block utilization of glucose imported by non-PTS pathways, we determined that of the two glucose transport pathways in GAS (PTS and non-PTS), the non-PTS pathway played a more significant role in glucose transport. However, the expression of both pathways is linked by a currently unknown mechanism, as blocking the non-PTS uptake of glucose reduces ptsI (EI) expression. Similar to the effects of the deletion of the PTS pathway, lack of the non-PTS pathway also leads to the early activity of Streptolysin S. However, this early activity did not adversely or favorably affect survival of ΔnagC in whole human blood. In a subcutaneous murine infection model, ΔnagC-infected mice showed increased lesion severity at the local site of infection; although, lesion size and dissemination from the site of infection was similar to wild type. Here, we show that glucose transport in GAS is primarily via a non-PTS pathway. The route of glucose transport differentially affects the survival of GAS in whole human blood, as well as the lesion size at the local site of infection in a murine skin infection model.
Collapse
Affiliation(s)
- Ganesh S Sundar
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, College Park, MD, United States
| | - Emrul Islam
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, College Park, MD, United States
| | - Rezia D Braza
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, College Park, MD, United States
| | - Aliyah B Silver
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, College Park, MD, United States
| | - Yoann Le Breton
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, College Park, MD, United States
| | - Kevin S McIver
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, College Park, MD, United States
| |
Collapse
|
27
|
Incremental Contributions of FbaA and Other Impetigo-Associated Surface Proteins to Fitness and Virulence of a Classical Group A Streptococcal Skin Strain. Infect Immun 2017; 85:IAI.00374-17. [PMID: 28808160 DOI: 10.1128/iai.00374-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 12/26/2022] Open
Abstract
Group A streptococci (GAS) are highly prevalent human pathogens whose primary ecological niche is the superficial epithelial layers of the throat and/or skin. Many GAS strains with a strong tendency to cause pharyngitis are distinct from strains that tend to cause impetigo; thus, genetic differences between them may confer host tissue-specific virulence. In this study, the FbaA surface protein gene was found to be present in most skin specialist strains but largely absent from a genetically related subset of pharyngitis isolates. In an ΔfbaA mutant constructed in the impetigo strain Alab49, loss of FbaA resulted in a slight but significant decrease in GAS fitness in a humanized mouse model of impetigo; the ΔfbaA mutant also exhibited decreased survival in whole human blood due to phagocytosis. In assays with highly sensitive outcome measures, Alab49ΔfbaA was compared to other isogenic mutants lacking virulence genes known to be disproportionately associated with classical skin strains. FbaA and PAM (i.e., the M53 protein) had additive effects in promoting GAS survival in whole blood. The pilus adhesin tip protein Cpa promoted Alab49 survival in whole blood and appears to fully account for the antiphagocytic effect attributable to pili. The finding that numerous skin strain-associated virulence factors make slight but significant contributions to virulence underscores the incremental contributions to fitness of individual surface protein genes and the multifactorial nature of GAS-host interactions.
Collapse
|
28
|
Itzek A, Chen Z, Merritt J, Kreth J. Effect of salivary agglutination on oral streptococcal clearance by human polymorphonuclear neutrophil granulocytes. Mol Oral Microbiol 2017; 32:197-210. [PMID: 27194631 PMCID: PMC5116291 DOI: 10.1111/omi.12164] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2016] [Indexed: 12/20/2022]
Abstract
Salivary agglutination is an important host defense mechanism to aggregate oral commensal bacteria as well as invading pathogens. Saliva flow and subsequent swallowing more easily clear aggregated bacteria compared with single cells. Phagocytic clearance of bacteria through polymorphonuclear neutrophil granulocytes also seems to increase to a certain extent with the size of bacterial aggregates. To determine a connection between salivary agglutination and the host innate immune response by phagocytosis, an in vitro agglutination assay was developed reproducing the average size of salivary bacterial aggregates. Using the oral commensal Streptococcus gordonii as a model organism, the effect of salivary agglutination on phagocytic clearance through polymorphonuclear neutrophil granulocytes was investigated. Here we describe how salivary aggregates of S. gordonii are readily cleared through phagocytosis, whereas single bacterial cells showed a significant delay in being phagocytosed and killed. Furthermore, before phagocytosis the polymorphonuclear neutrophil granulocytes were able to induce a specific de-aggregation, which was dependent on serine protease activity. The data presented suggest that salivary agglutination of bacterial cells leads to an ideal size for recognition by polymorphonuclear neutrophil granulocytes. As a first line of defense, these phagocytic cells are able to recognize the aggregates and de-aggregate them via serine proteases to a more manageable size for efficient phagocytosis and subsequent killing in the phagolysosome. This observed mechanism not only prevents the rapid spreading of oral bacterial cells while entering the bloodstream but would also avoid degranulation of involved polymorphonuclear neutrophil granulocytes, so preventing collateral damage to nearby tissue.
Collapse
Affiliation(s)
- Andreas Itzek
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zhiyun Chen
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Justin Merritt
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, OR, USA
| | - Jens Kreth
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
29
|
Vega LA, Valdes KM, Sundar GS, Belew AT, Islam E, Berge J, Curry P, Chen S, El-Sayed NM, Le Breton Y, McIver KS. The Transcriptional Regulator CpsY Is Important for Innate Immune Evasion in Streptococcus pyogenes. Infect Immun 2017; 85:e00925-16. [PMID: 27993974 PMCID: PMC5328483 DOI: 10.1128/iai.00925-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/13/2016] [Indexed: 01/01/2023] Open
Abstract
As an exclusively human pathogen, Streptococcus pyogenes (the group A streptococcus [GAS]) has specifically adapted to evade host innate immunity and survive in multiple tissue niches, including blood. GAS can overcome the metabolic constraints of the blood environment and expresses various immunomodulatory factors necessary for survival and immune cell resistance. Here we present our investigation of one such factor, the predicted LysR family transcriptional regulator CpsY. The encoding gene, cpsY, was initially identified as being required for GAS survival in a transposon-site hybridization (TraSH) screen in whole human blood. CpsY is homologous with transcriptional regulators of Streptococcus mutans (MetR), Streptococcus iniae (CpsY), and Streptococcus agalactiae (MtaR) that regulate methionine transport, amino acid metabolism, resistance to neutrophil-mediated killing, and survival in vivo Our investigation indicated that CpsY is involved in GAS resistance to innate immune cells of its human host. However, GAS CpsY does not manifest the in vitro phenotypes of its homologs in other streptococcal species. GAS CpsY appears to regulate a small set of genes that is markedly different from the regulons of its homologs. The differential expression of these genes depends on the growth medium, and CpsY modestly influences their expression. The GAS CpsY regulon includes known virulence factors (mntE, speB, spd, nga [spn], prtS [SpyCEP], and sse) and cell surface-associated factors of GAS (emm1, mur1.2, sibA [cdhA], and M5005_Spy0500). Intriguingly, the loss of CpsY in GAS does not result in virulence defects in murine models of infection, suggesting that CpsY function in immune evasion is specific to the human host.
Collapse
Affiliation(s)
- Luis A Vega
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kayla M Valdes
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Ganesh S Sundar
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Ashton T Belew
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Emrul Islam
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Jacob Berge
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Patrick Curry
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Steven Chen
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Najib M El-Sayed
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, USA
| | - Yoann Le Breton
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kevin S McIver
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
30
|
Sumioka R, Nakata M, Okahashi N, Li Y, Wada S, Yamaguchi M, Sumitomo T, Hayashi M, Kawabata S. Streptococcus sanguinis induces neutrophil cell death by production of hydrogen peroxide. PLoS One 2017; 12:e0172223. [PMID: 28222125 PMCID: PMC5319702 DOI: 10.1371/journal.pone.0172223] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/01/2017] [Indexed: 11/18/2022] Open
Abstract
Streptococcus is the dominant bacterial genus in the human oral cavity and a leading cause of infective endocarditis. Streptococcus sanguinis belongs to the mitis group of streptococci and produces hydrogen peroxide (H2O2) by the action of SpxB, a pyruvate oxidase. In this study, we investigated the involvement of SpxB in survival of S. sanguinis in human blood and whether bacterial H2O2 exhibits cytotoxicity against human neutrophils. Results of a bactericidal test with human whole blood revealed that the spxB mutation in S. sanguinis is detrimental to its survival in blood. When S. sanguinis strains were exposed to isolated neutrophils, the bacterial survival rate was significantly decreased by spxB deletion. Furthermore, human neutrophils exposed to the S. sanguinis wild-type strain, in contrast to those exposed to an spxB mutant strain, underwent cell death with chromatin de-condensation and release of web-like extracellular DNA, reflecting induction of neutrophil extracellular traps (NETs). Since reactive oxygen species-mediated NET induction requires citrullination of arginine residues in histone proteins and subsequent chromatin de-condensation, we examined citrullination levels of histone in infected neutrophils. It is important to note that the citrullinated histone H3 was readily detected in neutrophils infected with the wild-type strain, as compared to infection with the spxB mutant strain. Moreover, decomposition of streptococcal H2O2 with catalase reduced NET induction. These results suggest that H2O2 produced by S. sanguinis provokes cell death of neutrophils and NET formation, thus potentially affecting bacterial survival in the bloodstream.
Collapse
Affiliation(s)
- Ryuichi Sumioka
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Masanobu Nakata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
- * E-mail:
| | - Nobuo Okahashi
- Center for Frontier Oral Science, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Yixuan Li
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Satoshi Wada
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Masaya Yamaguchi
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Tomoko Sumitomo
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Mikako Hayashi
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| |
Collapse
|
31
|
Honda-Ogawa M, Sumitomo T, Mori Y, Hamd DT, Ogawa T, Yamaguchi M, Nakata M, Kawabata S. Streptococcus pyogenes Endopeptidase O Contributes to Evasion from Complement-mediated Bacteriolysis via Binding to Human Complement Factor C1q. J Biol Chem 2017; 292:4244-4254. [PMID: 28154192 DOI: 10.1074/jbc.m116.749275] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/18/2017] [Indexed: 01/14/2023] Open
Abstract
Streptococcus pyogenes secretes various virulence factors for evasion from complement-mediated bacteriolysis. However, full understanding of the molecules possessed by this organism that interact with complement C1q, an initiator of the classical complement pathway, remains elusive. In this study, we identified an endopeptidase of S. pyogenes, PepO, as an interacting molecule, and investigated its effects on complement immunity and pathogenesis. Enzyme-linked immunosorbent assay and surface plasmon resonance analysis findings revealed that S. pyogenes recombinant PepO bound to human C1q in a concentration-dependent manner under physiological conditions. Sites of inflammation are known to have decreased pH levels, thus the effects of PepO on bacterial evasion from complement immunity was analyzed in a low pH condition. Notably, under low pH conditions, PepO exhibited a higher affinity for C1q as compared with IgG, and PepO inhibited the binding of IgG to C1q. In addition, pepO deletion rendered S. pyogenes more susceptible to the bacteriocidal activity of human serum. Also, observations of the morphological features of the pepO mutant strain (ΔpepO) showed damaged irregular surfaces as compared with the wild-type strain (WT). WT-infected tissues exhibited greater severity and lower complement activity as compared with those infected by ΔpepO in a mouse skin infection model. Furthermore, WT infection resulted in a larger accumulation of C1q than that with ΔpepO. Our results suggest that interaction of S. pyogenes PepO with C1q interferes with the complement pathway, which enables S. pyogenes to evade complement-mediated bacteriolysis under acidic conditions, such as seen in inflammatory sites.
Collapse
Affiliation(s)
- Mariko Honda-Ogawa
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| | - Tomoko Sumitomo
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| | - Yasushi Mori
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry.,Division of Special Care Dentistry, Osaka University Dental Hospital, and
| | - Dalia Talat Hamd
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| | - Taiji Ogawa
- Department of Prosthodontics, Gerodontology and Oral Rehabilitation, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masaya Yamaguchi
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| | - Masanobu Nakata
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| | - Shigetada Kawabata
- From the Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry,
| |
Collapse
|
32
|
Courtney HS, Niedermeyer SE, Penfound TA, Hohn CM, Greeley A, Dale JB. Trivalent M-related protein as a component of next generation group A streptococcal vaccines. Clin Exp Vaccine Res 2017; 6:45-49. [PMID: 28168173 PMCID: PMC5292357 DOI: 10.7774/cevr.2017.6.1.45] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/21/2016] [Accepted: 01/02/2017] [Indexed: 11/15/2022] Open
Abstract
PURPOSE There is a need to broaden protective coverage of M protein-based vaccines against group A streptococci (GAS) because coverage of the current 30-valent M protein vaccine does not extend to all emm types. An additional GAS antigen and virulence factor that could potentially extend vaccine coverage is M-related protein (Mrp). Previous work indicated that there are three structurally related families of Mrp (MrpI, MrpII, and MrpIII) and peptides of all three elicited bactericidal antibodies against multiple emm types. The purpose of this study was to determine if a recombinant form containing Mrp from the three families would evoke bactericidal antiserum and to determine if this antiserum could enhance the effectiveness of antisera to the 30-valent M protein vaccine. MATERIALS AND METHODS A trivalent recombinant Mrp (trMrp) protein containing N-terminal fragments from the three families (trMrp) was constructed, purified and used to immunize rabbits. Anti-trMrp sera contained high titers of antibodies against the trMrp immunogen and recombinant forms representing MrpI, MrpII, and MrpIII. RESULTS The antisera opsonized emm types of GAS representing each Mrp family and also opsonized emm types not covered by the 30-valent M protein-based vaccine. Importantly, a combination of trMrp and 30-valent M protein antiserum resulted in higher levels of opsonization of GAS than either antiserum alone. CONCLUSION These findings suggest that trMrp may be an effective addition to future constructs of GAS vaccines.
Collapse
Affiliation(s)
- Harry S Courtney
- Department of Medicine, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA.; Department of Medicine, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Shannon E Niedermeyer
- Department of Medicine, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA.; Department of Medicine, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Thomas A Penfound
- Department of Medicine, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA.; Department of Medicine, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Claudia M Hohn
- Department of Medicine, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA.; Department of Medicine, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Adam Greeley
- Department of Medicine, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA.; Department of Medicine, Veterans Affairs Medical Center, Memphis, TN, USA
| | - James B Dale
- Department of Medicine, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA.; Department of Medicine, Veterans Affairs Medical Center, Memphis, TN, USA.; Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
33
|
Tsai JYC, Loh JMS, Clow F, Lorenz N, Proft T. The Group A Streptococcus serotype M2 pilus plays a role in host cell adhesion and immune evasion. Mol Microbiol 2016; 103:282-298. [PMID: 27741558 DOI: 10.1111/mmi.13556] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2016] [Indexed: 01/22/2023]
Abstract
Group A Streptococcus (GAS), or Streptococcus pyogenes, is a human pathogen that causes diseases ranging from skin and soft tissue infections to severe invasive diseases, such as toxic shock syndrome. Each GAS strain carries a particular pilus type encoded in the variable fibronectin-binding, collagen-binding, T antigen (FCT) genomic region. Here, we describe the functional analysis of the serotype M2 pilus encoded in the FCT-6 region. We found that, in contrast to other investigated GAS pili, the ancillary pilin 1 lacks adhesive properties. Instead, the backbone pilin is important for host cell adhesion and binds several host factors, including fibronectin and fibrinogen. Using a panel of recombinant pilus proteins, GAS gene deletion mutants and Lactococcus lactis gain-of-function mutants we show that, unlike other GAS pili, the FCT-6 pilus also contributes to immune evasion. This was demonstrated by a delay in blood clotting, increased intracellular survival of the bacteria in macrophages, higher bacterial survival rates in human whole blood and greater virulence in a Galleria mellonella infection model in the presence of fully assembled FCT-6 pili.
Collapse
Affiliation(s)
- Jia-Yun C Tsai
- Department of Molecular Medicine & Pathology, School of Medical Sciences.,Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences.,Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Fiona Clow
- Department of Molecular Medicine & Pathology, School of Medical Sciences
| | - Natalie Lorenz
- Department of Molecular Medicine & Pathology, School of Medical Sciences
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences.,Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
The fruRBA Operon Is Necessary for Group A Streptococcal Growth in Fructose and for Resistance to Neutrophil Killing during Growth in Whole Human Blood. Infect Immun 2016; 84:1016-1031. [PMID: 26787724 DOI: 10.1128/iai.01296-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/15/2016] [Indexed: 01/01/2023] Open
Abstract
Bacterial pathogens rely on the availability of nutrients for survival in the host environment. The phosphoenolpyruvate-phosphotransferase system (PTS) is a global regulatory network connecting sugar uptake with signal transduction. Since the fructose PTS has been shown to impact virulence in several streptococci, including the human pathogen Streptococcus pyogenes(the group A Streptococcus[GAS]), we characterized its role in carbon metabolism and pathogenesis in the M1T1 strain 5448. Growth in fructose as a sole carbon source resulted in 103 genes affected transcriptionally, where the frulocus (fruRBA) was the most induced. Reverse transcriptase PCR showed that fruRBA formed an operon which was repressed by FruR in the absence of fructose, in addition to being under carbon catabolic repression. Growth assays and carbon utilization profiles revealed that although the entire fruoperon was required for growth in fructose, FruA was the main transporter for fructose and also was involved in the utilization of three additional PTS sugars: cellobiose, mannitol, and N-acetyl-D-galactosamine. The inactivation of sloR, a fruA homolog that also was upregulated in the presence of fructose, failed to reveal a role as a secondary fructose transporter. Whereas the ability of both ΔfruR and ΔfruB mutants to survive in the presence of whole human blood or neutrophils was impaired, the phenotype was not reproduced in murine whole blood, and those mutants were not attenuated in a mouse intraperitoneal infection. Since the ΔfruA mutant exhibited no phenotype in the human or mouse assays, we propose that FruR and FruB are important for GAS survival in a human-specific environment.
Collapse
|
35
|
Abstract
Staphylococcus aureus is a leading pathogen in surgical site, intensive care unit, and skin infections, as well as healthcare-associated pneumonias. These infections are associated with an enormous burden of morbidity, mortality, and increase of hospital length of stay and patient cost. S. aureus is impressively fast in acquiring antibiotic resistance, and multidrug-resistant strains are a serious threat to human health. Due to resistance or insufficient effectiveness, antibiotics and bundle measures leave a tremendous unmet medical need worldwide. There are no licensed vaccines on the market despite the significant efforts done by public and private initiatives. Indeed, vaccines tested in clinical trials in the last two decades have failed to show efficacy. However, they targeted single antigens and contained no adjuvants and efficacy trials were performed in severely ill subjects. Herein, we provide a comprehensive evaluation of potential target populations for efficacy trials taking into account key factors such as population size, incidence of S. aureus infection, disease outcome, primary endpoints, as well as practical advantages and disadvantages. We describe the whole-blood assay as a potential surrogate of protection, and we show the link between phase III clinical trial data of failed vaccines with their preclinical observations. Finally, we give our perspective on how new vaccine formulations and clinical development approaches may lead to successful S. aureus vaccines.
Collapse
|
36
|
Sheel M, Moreland NJ, Fraser JD, Carapetis J. Development of Group A streptococcal vaccines: an unmet global health need. Expert Rev Vaccines 2015; 15:227-38. [DOI: 10.1586/14760584.2016.1116946] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Meru Sheel
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Nicole J Moreland
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - John D Fraser
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Jonathan Carapetis
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
- Princess Margaret Hospital for Children, Perth, Australia
| |
Collapse
|
37
|
Vivas-Alegre S, Fernández-Natal I, López-Fidalgo E, Rivero-Lezcano OM. Preparation of inocula for experimental infection of blood with Streptococcus pneumoniae. MethodsX 2015; 2:463-8. [PMID: 26844211 PMCID: PMC4688400 DOI: 10.1016/j.mex.2015.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 11/04/2015] [Indexed: 11/18/2022] Open
Abstract
Experimental infections of either cells or animals require the preparation of good quality inocula. Unfortunately, the important pulmonary pathogen Streptococcus pneumoniae is a fastidious microorganism that suffers an autolysis process when cultured in vitro. Supplementation of Todd–Hewitt broth with a biological buffer (20 mM Tris–HCl, pH = 7.8) promotes a six hours delay in the beginning of the autolysis process. Additional improvements include washing bacteria before freezing, avoiding manipulations after thawing, and the use of glycerol (<18%) as a cryoprotectant, instead of reagents like skimmed milk that may affect cell cultures. With the proposed protocol >70% of the frozen bacteria was viable after 28 weeks at −80 °C, and aliquots were highly homogeneous. We have tested their utility in a whole blood infection model and have found that human plasma exhibits a higher microbicidal activity than whole blood, a result that we have not found previously reported. Additionally, we have also observed significant variations in the antimicrobial activity against different strains, which might be related to their virulence.Media culture buffering extends S. pneumoniae viability for 6 h. Washing before freezing of single use aliquots minimizes manipulation after thawing. Experimental infection with the frozen inocula has shown that plasma has higher bactericidal activity than blood.
Collapse
Affiliation(s)
- Santiago Vivas-Alegre
- Servicio de Digestivo, Hospital de León, Altos de Nava s/n, 24008 León, Spain; Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | | | | | - Octavio Miguel Rivero-Lezcano
- Institute of Biomedicine (IBIOMED), University of León, León, Spain; Unidad de Investigación, Hospital de León, Altos de Nava s/n, 24008 León, Spain; Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Spain
| |
Collapse
|
38
|
Raz A, Tanasescu AM, Zhao AM, Serrano A, Alston T, Sol A, Bachrach G, Fischetti VA. Streptococcus pyogenes Sortase Mutants Are Highly Susceptible to Killing by Host Factors Due to Aberrant Envelope Physiology. PLoS One 2015; 10:e0140784. [PMID: 26484774 PMCID: PMC4617865 DOI: 10.1371/journal.pone.0140784] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/30/2015] [Indexed: 12/31/2022] Open
Abstract
Cell wall anchored virulence factors are critical for infection and colonization of the host by Gram-positive bacteria. Such proteins have an N-terminal leader sequence and a C-terminal sorting signal, composed of an LPXTG motif, a hydrophobic stretch, and a few positively charged amino acids. The sorting signal halts translocation across the membrane, allowing sortase to cleave the LPXTG motif, leading to surface anchoring. Deletion of sortase prevents the anchoring of virulence factors to the wall; the effects on bacterial physiology however, have not been thoroughly characterized. Here we show that deletion of Streptococcus pyogenes sortase A leads to accumulation of sorting intermediates, particularly at the septum, altering cellular morphology and physiology, and compromising membrane integrity. Such cells are highly sensitive to cathelicidin, and are rapidly killed in blood and plasma. These phenomena are not a loss-of-function effect caused by the absence of anchored surface proteins, but specifically result from the accumulation of sorting intermediates. Reduction in the level of sorting intermediates leads to a return of the sortase mutant to normal morphology, while expression of M protein with an altered LPXTG motif in wild type cells leads to toxicity in the host environment, similar to that observed in the sortase mutant. These unanticipated effects suggest that inhibition of sortase by small-molecule inhibitors could similarly lead to the rapid elimination of pathogens from an infected host, making such inhibitors much better anti-bacterial agents than previously believed.
Collapse
Affiliation(s)
- Assaf Raz
- Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230 York Avenue Box 172, New York, New York, 10065, United States of America
| | - Ana-Maria Tanasescu
- Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230 York Avenue Box 172, New York, New York, 10065, United States of America
| | - Anna M. Zhao
- Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230 York Avenue Box 172, New York, New York, 10065, United States of America
| | - Anna Serrano
- Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230 York Avenue Box 172, New York, New York, 10065, United States of America
| | - Tricia Alston
- Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230 York Avenue Box 172, New York, New York, 10065, United States of America
| | - Asaf Sol
- Institute of Dental Sciences, Hebrew University - Hadassah School of Dental Medicine, Jerusalem 91120, Israel
| | - Gilad Bachrach
- Institute of Dental Sciences, Hebrew University - Hadassah School of Dental Medicine, Jerusalem 91120, Israel
| | - Vincent A. Fischetti
- Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230 York Avenue Box 172, New York, New York, 10065, United States of America
| |
Collapse
|
39
|
Kim YT, Kim SK, Jeon YJ, Park SJ. Seahorse-derived peptide suppresses invasive migration of HT1080 fibrosarcoma cells by competing with intracellular α-enolase for plasminogen binding and inhibiting uPA-mediated activation of plasminogen. BMB Rep 2015; 47:691-6. [PMID: 24602611 PMCID: PMC4345514 DOI: 10.5483/bmbrep.2014.47.12.235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Indexed: 01/22/2023] Open
Abstract
α-Enolase is a glycolytic enzyme and a surface receptor for plasminogen. α-Enolase-bound plasminogen promotes tumor cell invasion and cancer metastasis by activating plasmin and consequently degrading the extracellular matrix degradation. Therefore, α-enolase and plasminogen are novel targets for cancer therapy. We found that the amino acid sequence of a peptide purified from enzymatic hydrolysates of seahorse has striking similarities to that of α-enolase. In this study, we report that this peptide competes with cellular α-enolase for plasminogen binding and suppresses urokinase plasminogen activator (uPA)-mediated activation of plasminogen, which results in decreased invasive migration of HT1080 fibrosarcoma cells. In addition, the peptide treatment decreased the expression levels of uPA compared to that of untreated controls. These results provide new insight into the mechanism by which the seahorse-derived peptide suppresses invasive properties of human cancer cells. Our findings suggest that this peptide could emerge as a potential therapeutic agent for cancer. [BMB Reports 2014; 47(12): 691-696]
Collapse
Affiliation(s)
- Yong-Tae Kim
- Department of Food Science and Technology, Kunsan National University, Kunsan 573-701, Korea
| | - Se-kwon Kim
- Department of Chemistry, Pukyong National University, Busan 608-737, Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Korea
| | - Sun Joo Park
- Department of Chemistry, Pukyong National University, Busan 608-737, Korea
| |
Collapse
|
40
|
Lannergård J, Kristensen BM, Gustafsson MCU, Persson JJ, Norrby-Teglund A, Stålhammar-Carlemalm M, Lindahl G. Sequence variability is correlated with weak immunogenicity in Streptococcus pyogenes M protein. Microbiologyopen 2015; 4:774-89. [PMID: 26175306 PMCID: PMC4618610 DOI: 10.1002/mbo3.278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/18/2015] [Indexed: 11/06/2022] Open
Abstract
The M protein of Streptococcus pyogenes, a major bacterial virulence factor, has an amino-terminal hypervariable region (HVR) that is a target for type-specific protective antibodies. Intriguingly, the HVR elicits a weak antibody response, indicating that it escapes host immunity by two mechanisms, sequence variability and weak immunogenicity. However, the properties influencing the immunogenicity of regions in an M protein remain poorly understood. Here, we studied the antibody response to different regions of the classical M1 and M5 proteins, in which not only the HVR but also the adjacent fibrinogen-binding B repeat region exhibits extensive sequence divergence. Analysis of antisera from S. pyogenes-infected patients, infected mice, and immunized mice showed that both the HVR and the B repeat region elicited weak antibody responses, while the conserved carboxy-terminal part was immunodominant. Thus, we identified a correlation between sequence variability and weak immunogenicity for M protein regions. A potential explanation for the weak immunogenicity was provided by the demonstration that protease digestion selectively eliminated the HVR-B part from whole M protein-expressing bacteria. These data support a coherent model, in which the entire variable HVR-B part evades antibody attack, not only by sequence variability but also by weak immunogenicity resulting from protease attack.
Collapse
Affiliation(s)
- Jonas Lannergård
- Department of Laboratory Medicine, Lund University, Lund, Sweden.,Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg C, Denmark
| | | | | | - Jenny J Persson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Karolinska Institutet, Huddinge University Hospital, Stockholm, Sweden
| | | | - Gunnar Lindahl
- Department of Laboratory Medicine, Lund University, Lund, Sweden.,Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
41
|
Correlates of Protection for M Protein-Based Vaccines against Group A Streptococcus. J Immunol Res 2015; 2015:167089. [PMID: 26101780 PMCID: PMC4458553 DOI: 10.1155/2015/167089] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/28/2015] [Accepted: 05/03/2015] [Indexed: 11/17/2022] Open
Abstract
Group A streptococcus (GAS) is known to cause a broad spectrum of illness, from pharyngitis and impetigo, to autoimmune sequelae such as rheumatic heart disease, and invasive diseases. It is a significant cause of infectious disease morbidity and mortality worldwide, but no efficacious vaccine is currently available. Progress in GAS vaccine development has been hindered by a number of obstacles, including a lack of standardization in immunoassays and the need to define human correlates of protection. In this review, we have examined the current immunoassays used in both GAS and other organisms, and explored the various challenges in their implementation in order to propose potential future directions to identify a correlate of protection and facilitate the development of M protein-based vaccines, which are currently the main GAS vaccine candidates.
Collapse
|
42
|
Pozzi C, Lofano G, Mancini F, Soldaini E, Speziale P, De Gregorio E, Rappuoli R, Bertholet S, Grandi G, Bagnoli F. Phagocyte subsets and lymphocyte clonal deletion behind ineffective immune response to Staphylococcus aureus. FEMS Microbiol Rev 2015; 39:750-63. [PMID: 25994610 DOI: 10.1093/femsre/fuv024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2015] [Indexed: 01/14/2023] Open
Abstract
Lack of known mechanisms of protection against Staphylococcus aureus in humans is hindering development of efficacious vaccines. Preclinical as well as clinical data suggest that antibodies play an important role against S. aureus. For instance, certain hypogammaglobulinaemic patients are at increased risk of staphylococcal infections. However, development of effective humoral response may be dampened by converging immune-evasion mechanisms of S. aureus. We hypothesize that B-cell proliferation induced by staphylococcal protein A (SpA) and continuous antigen exposure, without the proper T-cell help and cytokine stimuli, leads to antigen-activated B-cell deletion and anergy. Recent findings suggest an important role of type I neutrophils (PMN-I) and conventionally activated macrophages (M1) against S. aureus, while alternatively activated macrophages (M2) favour biofilm persistence and sepsis. In addition, neutrophil-macrophage cooperation promotes extravasation and activation of neutrophils as well as clearance of bacteria ensnared in neutrophil extracellular traps. Activation of these processes is modulated by cytokines and T cells. Indeed, low CD4(+) T-cell counts represent an important risk factor for skin infections and bacteraemia in patients. Altogether, these observations could lead to the identification of predictive correlates of protection and ways for shifting the balance of the response to the benefit of the host through vaccination.
Collapse
Affiliation(s)
- Clarissa Pozzi
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | - Giuseppe Lofano
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | - Francesca Mancini
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | | | - Pietro Speziale
- Department of Molecular Medicine, Institute of Biochemistry, 27100 Pavia, Italy
| | - Ennio De Gregorio
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | - Rino Rappuoli
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | - Sylvie Bertholet
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | - Guido Grandi
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| | - Fabio Bagnoli
- Novartis Vaccines, Research Center, via Fiorentina 1, 53100 Siena, Italy
| |
Collapse
|
43
|
Zhang Y, Okada R, Isaka M, Tatsuno I, Isobe KI, Hasegawa T. Analysis of the roles of NrdR and DnaB from Streptococcus pyogenes in response to host defense. APMIS 2014; 123:252-9. [PMID: 25469586 DOI: 10.1111/apm.12340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/01/2014] [Indexed: 12/01/2022]
Abstract
Toxic shock syndrome caused by Streptococcus pyogenes (S. pyogenes) is a re-emerging infectious disease. Many virulence-associated proteins play important roles in its pathogenesis and the production of these proteins is controlled by many regulatory factors. CovS is one of the most important two-component sensor proteins in S. pyogenes, and it has been analyzed extensively. Our recent analyses revealed the existence of a transposon between covS and nrdR in several strains, and we speculated that this insertion has some importance. Hence, we examined the significances of the NrdR stand-alone regulator and DnaB, which is encoded by the gene located immediately downstream of nrdR in S. pyogenes infection. We established an nrdR-only knockout strain, and both nrdR and partial dnaB knockout strain. These established knockout strains exhibited a deteriorated response to H2 O2 exposure. nrdR and partial dnaB knockout strain was more easily killed by human polynuclear blood cells, but the nrdR-only knockout strain had no significant difference compared to wild type in contrast to the combined knockout strain. In addition, the mouse infection model experiment illustrated that nrdR and partial dnaB knockout strain, but not the nrdR-only knockout strain, was less virulent compared with the parental strain. These results suggest that DnaB is involved in response to host defense.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Bacteriology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Asymptomatic carriage of group A streptococcus is associated with elimination of capsule production. Infect Immun 2014; 82:3958-67. [PMID: 25024363 DOI: 10.1128/iai.01788-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Humans commonly carry pathogenic bacteria asymptomatically, but despite decades of study, the underlying molecular contributors remain poorly understood. Here, we show that a group A streptococcus carriage strain contains a frameshift mutation in the hasA gene resulting in loss of hyaluronic acid capsule biosynthesis. This mutation was repaired by allelic replacement, resulting in restoration of capsule production in the isogenic derivative strain. The "repaired" isogenic strain was significantly more virulent than the carriage strain in a mouse model of necrotizing fasciitis and had enhanced growth ex vivo in human blood. Importantly, the repaired isogenic strain colonized the mouse oropharynx with significantly greater bacterial burden and had significantly reduced ability to internalize into cultured epithelial cells than the acapsular carriage strain. We conducted full-genome sequencing of 81 strains cultured serially from 19 epidemiologically unrelated human subjects and discovered the common theme that mutations negatively affecting capsule biosynthesis arise in vivo in the has operon. The significantly decreased capsule production is a key factor contributing to the molecular détente between pathogen and host. Our discoveries suggest a general model for bacterial pathogens in which mutations that downregulate or ablate virulence factor production contribute to carriage.
Collapse
|
45
|
Ma C, Liu Z, Li W, Qian X, Zhang S, Gao X, Jiang S, Wei L. FbaA- and M protein-based multi-epitope vaccine elicits strong protective immune responses against group A streptococcus in mouse model. Microbes Infect 2014; 16:409-18. [PMID: 24704476 DOI: 10.1016/j.micinf.2014.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/20/2014] [Accepted: 03/20/2014] [Indexed: 11/25/2022]
Abstract
We report the construction of a recombinant multivalent vaccine against group A streptococcus (GAS), designated F7M5. It contains seven predominant epitopes of FbaA identified by phage display technology, five non-tissue cross-reactive M protein fragments expressed on four selected serotypes prevalent in China, a Trojan antigen (TA) and a poly-alanine DR epitope (PADRE). BALB/c mice were immunized subcutaneously with F7M5 formulated with Freund's adjuvant, using recombinant FbaA and M protein in parallel as control. Using enzyme-linked immunosorbent assay (ELISA), mouse immune sera were assayed for IgG titers, IgG subclasses, and binding of F7M5 with M1GAS. Results indicated that the multivalent vaccine was highly immunogenic and elicited a balanced IgG1/IgG2a response. We also tested the reactivity of F7M5 to antistreptolysin O (ASO) antibodies in sera of GAS-infected patients and found a 95.8% positive rate, indicating that the epitopes of the vaccine were widely expressed in the prevalent serotypes of GAS. More importantly, the F7M5 vaccine elicited strong protective immune responses against lethal-dose challenge with a survival rate of 90%, but induced no cross-reactions or pathological lesions in mouse model, suggesting that F7M5 can be further developed as an effective and safe anti-GAS vaccine.
Collapse
Affiliation(s)
- Cuiqing Ma
- Department of Immunology, Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Zheng Liu
- Department of Immunology, Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Wenjian Li
- Department of Immunology, Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Xuesong Qian
- Department of Immunology, Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Song Zhang
- Third Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Xue Gao
- Department of Immunology, Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China.
| | - Lin Wei
- Department of Immunology, Basic Medical College, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
46
|
Gera K, McIver KS. Laboratory growth and maintenance of Streptococcus pyogenes (the Group A Streptococcus, GAS). ACTA ACUST UNITED AC 2013; 30:9D.2.1-9D.2.13. [PMID: 24510893 DOI: 10.1002/9780471729259.mc09d02s30] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Streptococcus pyogenes is a Gram-positive bacterium that strictly infects humans. It is the causative agent of a broad spectrum of diseases accounting for millions of infections and at least 517,000 deaths each year worldwide. It is a nutritionally fastidious organism that ferments sugars to produce lactic acid and has strict requirements for growth. To aid in the study of this organism, this unit describes the growth and maintenance of S. pyogenes.
Collapse
Affiliation(s)
- Kanika Gera
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Kevin S McIver
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| |
Collapse
|
47
|
Natural variation in the promoter of the gene encoding the Mga regulator alters host-pathogen interactions in group a Streptococcus carrier strains. Infect Immun 2013; 81:4128-38. [PMID: 23980109 DOI: 10.1128/iai.00405-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Humans commonly carry pathogenic bacteria asymptomatically, but the molecular factors underlying microbial asymptomatic carriage are poorly understood. We previously reported that two epidemiologically unassociated serotype M3 group A Streptococcus (GAS) carrier strains had an identical 12-bp deletion in the promoter of the gene encoding Mga, a global positive gene regulator. Herein, we report on studies designed to test the hypothesis that the identified 12-bp deletion in the mga promoter alters GAS virulence, thereby potentially contributing to the asymptomatic carrier phenotype. Using allelic exchange, we introduced the variant promoter into a serotype M3 invasive strain and the wild-type promoter into an asymptomatic carrier strain. Compared to strains with the wild-type mga promoter, we discovered that strains containing the promoter with the 12-bp deletion produced significantly fewer mga and Mga-regulated gene transcripts. Consistent with decreased mga transcripts, strains containing the variant mga promoter were also significantly less virulent in in vivo and ex vivo models of GAS disease. Further, we provide evidence that the pleiotropic regulator protein CodY binds to the mga promoter and that the 12-bp deletion in the mga promoter reduces CodY-mediated mga transcription. We conclude that the naturally occurring 12-bp deletion in the mga promoter significantly alters the pathogen-host interaction of these asymptomatic carrier strains. Our findings provide new insight into the molecular basis of the carrier state of an important human pathogen.
Collapse
|
48
|
Good MF, Batzloff MR, Pandey M. Strategies in the development of vaccines to prevent infections with group A streptococcus. Hum Vaccin Immunother 2013; 9:2393-7. [PMID: 23863455 DOI: 10.4161/hv.25506] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
There has long been interest and demand for the development of a vaccine to prevent infections caused by the Gram-positive organism group A streptococcus. Despite numerous efforts utilizing advanced approaches such as genomics, proteomics and bio-informatics, there is currently no vaccine. Here we review various strategies employed to achieve this goal. We also discuss the approach that we have pursued, a non-host reactive, conformationally constrained minimal B cell epitope from within the C-repeat region of M-protein, and the potential limitations in moving forward.
Collapse
Affiliation(s)
- Michael F Good
- Institute for Glycomics; Griffith University, Gold Coast campus; QLD Australia
| | | | | |
Collapse
|
49
|
Honda-Ogawa M, Ogawa T, Terao Y, Sumitomo T, Nakata M, Ikebe K, Maeda Y, Kawabata S. Cysteine proteinase from Streptococcus pyogenes enables evasion of innate immunity via degradation of complement factors. J Biol Chem 2013; 288:15854-64. [PMID: 23589297 DOI: 10.1074/jbc.m113.469106] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Streptococcus pyogenes is an important human pathogen that causes invasive diseases such as necrotizing fasciitis, sepsis, and streptococcal toxic shock syndrome. We investigated the function of a major cysteine protease from S. pyogenes that affects the amount of C1-esterase inhibitor (C1-INH) and other complement factors and aimed to elucidate the mechanism involved in occurrence of streptococcal toxic shock syndrome from the aspect of the complement system. First, we revealed that culture supernatant of a given S. pyogenes strain and recombinant SpeB degraded the C1-INH. Then, we determined the N-terminal sequence of the C1-INH fragment degraded by recombinant SpeB. Interestingly, the region containing one of the identified cleavage sites is not present in patients with C1-INH deficiency. Scanning electron microscopy of the speB mutant incubated in human serum showed the abnormal superficial architecture and irregular oval structure. Furthermore, unlike the wild-type strain, that mutant strain showed lower survival capacity than normal as compared with heat-inactivated serum, whereas it had a significantly higher survival rate in serum without the C1-INH than in normal serum. Also, SpeB degraded multiple complement factors and the membrane attack complex. Flow cytometric analyses revealed deposition of C9, one of the components of membrane the attack complex, in greater amounts on the surface of the speB mutant, whereas lower amounts of C9 were bound to the wild-type strain surface. These results suggest that SpeB can interrupt the human complement system via degrading the C1-INH, thus enabling S. pyogenes to evade eradication in a hostile environment.
Collapse
Affiliation(s)
- Mariko Honda-Ogawa
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, 1-8, Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ding Y, Ni Q, Liu J, Yu B. Immunogenicity of a divalent group A streptococcal vaccine. Rheumatol Int 2013; 33:1013-20. [PMID: 22872049 DOI: 10.1007/s00296-012-2455-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 07/07/2012] [Indexed: 11/29/2022]
Abstract
We designed and recombined the polypeptide based on the M protein of group A streptococci (GAS)--the causative pathogen of rheumatic fever and rheumatic heart disease, which would be a divalent vaccine to prevent and defend the diseases in relation to the different GAS strains. A divalent vaccine comprising three different peptide epitopes of the antiphagocytic M protein of GAS--an aminoterminal specific sequences, respectively, from the M1 and M12 proteins and J14 peptide (ASREAKKQVEKALE) within the highly conserved C-terminal repeat region of the M1 and M12 proteins--was subcutaneously delivered to mice with the adjuvant. Furthermore, the antisera titers of mice inoculated with the divalent vaccine were assayed by ELISA, and then opsonization and percentage killing against two different GAS serotypes were completed. Our data demonstrated that antisera raised against the divalent vaccine containing amino acids and M-protein-conserved C repeat region are able to kill several GAS strains isolated from the Guangzhou population. Therefore, the divalent vaccine can be used to prevent those diseases caused by GAS in an endemic area. We successfully construct the M-protein-based divalent vaccine that can bring out a high-level antisera titer of mice vaccinated with it. So, the vaccine has the potential to be used to prevent diseases caused by GAS in our country.
Collapse
Affiliation(s)
- Yuexia Ding
- Department of Cardiology, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510700, China
| | | | | | | |
Collapse
|