1
|
de Crombrugghe G, Schiavolin L, Osowicki J, Steer AC, Botteaux A, Smeesters PR. M1 and done? Global assessment of the invasive potential of group A streptococcal strains. THE LANCET. MICROBE 2025:101123. [PMID: 40328277 DOI: 10.1016/j.lanmic.2025.101123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 05/08/2025]
Affiliation(s)
- Gabrielle de Crombrugghe
- Molecular Bacteriology, European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium; Department of Paediatrics, Brussels University Hospital, Academic Children Hospital Queen Fabiola, Université libre de Bruxelles, Brussels 1020, Belgium.
| | - Lionel Schiavolin
- Molecular Bacteriology, European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Joshua Osowicki
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Department of Infectious Diseases, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Andrew C Steer
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Department of Infectious Diseases, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Anne Botteaux
- Molecular Bacteriology, European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Pierre R Smeesters
- Molecular Bacteriology, European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium; Department of Paediatrics, Brussels University Hospital, Academic Children Hospital Queen Fabiola, Université libre de Bruxelles, Brussels 1020, Belgium; Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Brouwer S, Das S, Hayes AJ, Bertolla OM, Davies MR, Walker MJ, Whiley DM, Irwin AD, Tickner JA. A Rapid Molecular Detection Tool for Toxigenic M1UK Streptococcus pyogenes. J Infect Dis 2025; 231:e375-e384. [PMID: 39206960 PMCID: PMC11841628 DOI: 10.1093/infdis/jiae437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/07/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The gradual replacement of the Streptococcus pyogenes M1global genotype by a newly emergent M1UK variant is a global public health threat warranting increased surveillance. M1UK differs from progenitor M1global genotype by 27 single-nucleotide polymorphisms and is characterized by increased speA superantigen expression in vitro. METHODS An allele-specific real-time polymerase chain reaction assay was developed for the rapid detection of M1UK strains. The assay was used in combination with whole genome sequencing to determine emm (sub)type distribution for 51 invasive (n = 9) and noninvasive (n = 42) S pyogenes clinical isolates. RESULTS Emm1 was the most prevalent S pyogenes emm serotype (n = 11) in this set of clinical isolates, with M1UK being the dominant emm1 genotype (4/5 invasive, 3/6 noninvasive isolates). The assay accurately detected M1UK strains. Whole genome sequencing revealed continued presence of Australian M1UK sublineages associated with epidemic scarlet fever-causing S pyogenes in Asia. CONCLUSIONS Our study establishes a suitable target for detection of the toxigenic M1UK and confirms the maintenance of M1UK strains in Queensland, Australia. This assay can be deployed in laboratories and provides a valuable, cost-effective tool to enhance surveillance of the expanding M1UK clone.
Collapse
Affiliation(s)
- Stephan Brouwer
- Australian Infectious Diseases Research Centre and Institute for Molecular Bioscience
| | - Swairindhree Das
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane
| | - Andrew J Hayes
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria
| | - Olivia M Bertolla
- Australian Infectious Diseases Research Centre and Institute for Molecular Bioscience
| | - Mark R Davies
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria
| | - Mark J Walker
- Australian Infectious Diseases Research Centre and Institute for Molecular Bioscience
| | - David M Whiley
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane
- Queensland Paediatric Infectious Diseases Sakzewski Laboratory, Centre for Children's Health Research
| | - Adam D Irwin
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane
- Queensland Paediatric Infectious Diseases Sakzewski Laboratory, Centre for Children's Health Research
- Infection Management and Prevention Service, Queensland Children's Hospital, Brisbane, Australia
| | - Jacob A Tickner
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane
- Queensland Paediatric Infectious Diseases Sakzewski Laboratory, Centre for Children's Health Research
| |
Collapse
|
3
|
Richter J, Cork AJ, Ong Y, Keller N, Hayes AJ, Schembri MA, Jennison AV, Davies MR, Schroder K, Walker MJ, Brouwer S. Characterization of a novel covS SNP identified in Australian group A Streptococcus isolates derived from the M1 UK lineage. mBio 2025; 16:e0336624. [PMID: 39688411 PMCID: PMC11796353 DOI: 10.1128/mbio.03366-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Group A Streptococcus (GAS) is a human-adapted pathogen responsible for a variety of diseases. The GAS M1UK lineage has contributed significantly to the recently reported increases in scarlet fever and invasive infections. However, the basis for its evolutionary success is not yet fully understood. During the transition to systemic disease, the M1 serotype is known to give rise to spontaneous mutations in the control of virulence two-component regulatory system (CovRS) that confer a fitness advantage during invasive infections. Mutations that inactivate CovS function result in the de-repression of key GAS virulence factors such as streptolysin O (SLO), a pore-forming toxin and major trigger of inflammasome/interleukin-1β-dependent inflammation. Conversely, expression of the streptococcal cysteine protease SpeB, which is required during initial stages of colonization and onset of invasive disease, is typically lost in such mutants. In this study, we identified and characterized a novel covS single nucleotide polymorphism detected in three separate invasive M1UK isolates. The resulting CovSAla318Val mutation caused a significant upregulation of SLO resulting in increased inflammasome activation in human THP-1 macrophages, indicating an enhanced inflammatory potential. Surprisingly, SpeB production was unaffected. Site-directed mutagenesis was performed to assess the impact of this mutation on virulence and global gene expression. We found that the CovSAla318Val mutation led to subtle, virulence-specific changes of the CovRS regulon compared to previously characterized covS mutations, highlighting an unappreciated level of complexity in CovRS-dependent gene regulation. Continued longitudinal surveillance is warranted to determine whether this novel covS mutation will expand in the M1UK lineage.IMPORTANCEThe M1UK lineage of GAS has contributed to a recent global upsurge in scarlet fever and invasive infections. Understanding how GAS can become more virulent is critical for infection control and identifying new treatment approaches. The two-component CovRS system, comprising the sensor kinase CovS and transcription factor CovR, is a central regulator of GAS virulence genes. In the M1 serotype, covRS mutations are associated with an invasive phenotype. Such mutations have not been fully characterized in the M1UK lineage. This study identified a novel covS mutation in invasive Australian M1UK isolates that resulted in a more nuanced virulence gene regulation compared to previously characterized covS mutations. A representative isolate displayed upregulated SLO production and triggered amplified interleukin-1β secretion in infected human macrophages, indicating an enhanced inflammatory potential. These findings underscore the need for comprehensive analyses of covRS mutants to fully elucidate their contribution to M1UK virulence and persistence.
Collapse
Affiliation(s)
- Johanna Richter
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Amanda J. Cork
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Yvette Ong
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Nadia Keller
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Andrew J. Hayes
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Mark A. Schembri
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Amy V. Jennison
- Public and Environmental Health, Pathology Queensland, Queensland Health, Coopers Plains, Queensland, Australia
| | - Mark R. Davies
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience, Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark J. Walker
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Stephan Brouwer
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
4
|
Davis K, Abo YN, Steer AC, Osowicki J. Chains of misery: surging invasive group A streptococcal disease. Curr Opin Infect Dis 2024; 37:485-493. [PMID: 39259691 DOI: 10.1097/qco.0000000000001064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
PURPOSE OF REVIEW We describe the epidemiology of the recent global surge in invasive group A streptococcal (GAS) disease and consider its proximate and distal causes. We highlight important knowledge gaps regarding clinical management and discuss potential strategies for prevention. RECENT FINDINGS Rates of invasive GAS (iGAS) disease were increasing globally prior to the COVID-19 pandemic. Since mid-2022, following the worst years of the pandemic in 2020 and 2021, many countries with systems to monitor GAS syndromes have reported surges in cases of iGAS concurrent with increased scarlet fever, pharyngitis, and viral co-infections. The emergence of the hypervirulent M1 UK strain as a cause of iGAS, particularly in high income countries, is concerning. New data are emerging on the transmission dynamics of GAS. GAS remains universally susceptible to penicillin but there are increasing reports of macrolide and lincosamide resistance, particularly in invasive isolates, with uncertain clinical consequences. Intravenous immunoglobulin is used widely for streptococcal toxic shock syndrome and necrotizing soft tissue infections, although there is limited clinical evidence, and none from a completed randomized controlled trial. Intensive and expensive efforts at population-level control of GAS infections and postinfectious autoimmune complications have been only partially successful. The great hope for control of GAS diseases remains vaccine development. However, all modern vaccine candidates remain in the early development stage. SUMMARY In many countries, iGAS rates surged from mid-2022 in the aftermath of pandemic control measures and physical distancing. The emergence of a dominant hypervirulent strain is an important but incomplete explanation for this phenomenon. Clinical management of iGAS remains highly empirical and new data has not emerged. A vaccine remains the most likely means of achieving a sustainable reduction in the burden of iGAS.
Collapse
Affiliation(s)
- Kimberly Davis
- Tropical Diseases Research Group, Murdoch Children's Research Institute
- Department of Infection and Immunity, Monash Children's Hospital
- Department of Paediatrics, University of Melbourne
| | - Yara-Natalie Abo
- Tropical Diseases Research Group, Murdoch Children's Research Institute
- Department of Paediatrics, University of Melbourne
- Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital, Melbourne, Australia
| | - Andrew C Steer
- Tropical Diseases Research Group, Murdoch Children's Research Institute
- Department of Paediatrics, University of Melbourne
- Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital, Melbourne, Australia
| | - Joshua Osowicki
- Tropical Diseases Research Group, Murdoch Children's Research Institute
- Department of Paediatrics, University of Melbourne
- Infectious Diseases Unit, Department of General Medicine, Royal Children's Hospital, Melbourne, Australia
| |
Collapse
|
5
|
Bjånes E, Stream A, Janssen AB, Gibson PS, Bravo AM, Dahesh S, Baker JL, Varble A, Nizet V, Veening JW. An efficient in vivo-inducible CRISPR interference system for group A Streptococcus genetic analysis and pathogenesis studies. mBio 2024; 15:e0084024. [PMID: 38953375 PMCID: PMC11323564 DOI: 10.1128/mbio.00840-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
While genome-wide transposon mutagenesis screens have identified numerous essential genes in the significant human pathogen Streptococcus pyogenes (group A Streptococcus or GAS), many of their functions remain elusive. This knowledge gap is attributed in part to the limited molecular toolbox for controlling GAS gene expression and the bacterium's poor genetic transformability. CRISPR interference (CRISPRi), using catalytically inactive GAS Cas9 (dCas9), is a powerful approach to specifically repress gene expression in both bacteria and eukaryotes, but ironically, it has never been harnessed for controlled gene expression in GAS. In this study, we present a highly transformable and fully virulent serotype M1T1 GAS strain and introduce a doxycycline-inducible CRISPRi system for efficient repression of bacterial gene expression. We demonstrate highly efficient, oligo-based single guide RNA cloning directly to GAS, enabling the construction of a gene knockdown strain in just 2 days, in contrast to the several weeks typically required. The system is shown to be titratable and functional both in vitro and in vivo using a murine model of GAS infection. Furthermore, we provide direct in vivo evidence that the expression of the conserved cell division gene ftsZ is essential for GAS virulence, highlighting its promise as a target for emerging FtsZ inhibitors. Finally, we introduce SpyBrowse (https://veeninglab.com/SpyBrowse), a comprehensive and user-friendly online resource for visually inspecting and exploring GAS genetic features. The tools and methodologies described in this work are poised to facilitate fundamental research in GAS, contribute to vaccine development, and aid in the discovery of antibiotic targets. IMPORTANCE While group A Streptococcus (GAS) remains a predominant cause of bacterial infections worldwide, there are limited genetic tools available to study its basic cell biology. Here, we bridge this gap by creating a highly transformable, fully virulent M1T1 GAS strain. In addition, we established a tight and titratable doxycycline-inducible system and developed CRISPR interference (CRISPRi) for controlled gene expression in GAS. We show that CRISPRi is functional in vivo in a mouse infection model. Additionally, we present SpyBrowse, an intuitive and accessible genome browser (https://veeninglab.com/SpyBrowse). Overall, this work overcomes significant technical challenges of working with GAS and, together with SpyBrowse, represents a valuable resource for researchers in the GAS field.
Collapse
Affiliation(s)
- Elisabet Bjånes
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Alexandra Stream
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Axel B. Janssen
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Paddy S. Gibson
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Afonso M. Bravo
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Samira Dahesh
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Jonathon L. Baker
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, California, USA
| | - Andrew Varble
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Jan-Willem Veening
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6
|
Odo CM, Vega LA, Mukherjee P, DebRoy S, Flores AR, Shelburne SA. Emergent emm4 group A Streptococcus evidences a survival strategy during interaction with immune effector cells. Infect Immun 2024; 92:e0015224. [PMID: 38888310 PMCID: PMC11238559 DOI: 10.1128/iai.00152-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
The major gram-positive pathogen group A Streptococcus (GAS) is a model organism for studying microbial epidemics as it causes waves of infections. Since 1980, several GAS epidemics have been ascribed to the emergence of clones producing increased amounts of key virulence factors such as streptolysin O (SLO). Herein, we sought to identify mechanisms underlying our recently identified temporal clonal emergence among emm4 GAS, given that emergent strains did not produce augmented levels of virulence factors relative to historic isolates. By creating and analyzing isoallelic strains, we determined that a conserved mutation in a previously undescribed gene encoding a putative carbonic anhydrase was responsible for the defective in vitro growth observed in the emergent strains. We also identified that the emergent strains survived better inside macrophages and killed macrophages at lower rates than the historic strains. Via the creation of isogenic mutant strains, we linked the emergent strain "survival" phenotype to the downregulation of the SLO encoding gene and upregulation of the msrAB operon which encodes proteins involved in defense against extracellular oxidative stress. Our findings are in accord with recent surveillance studies which found a high ratio of mucosal (i.e., pharyngeal) relative to invasive infections among emm4 GAS. Since ever-increasing virulence is unlikely to be evolutionarily advantageous for a microbial pathogen, our data further understanding of the well-described oscillating patterns of virulent GAS infections by demonstrating mechanisms by which emergent strains adapt a "survival" strategy to outcompete previously circulating isolates.
Collapse
Affiliation(s)
- Chioma M. Odo
- Microbiology and Infectious Disease, MD Anderson UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Luis A. Vega
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Piyali Mukherjee
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sruti DebRoy
- Department of Infectious Disease, MD Anderson Cancer Center, Houston, Texas, USA
| | - Anthony R. Flores
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, University of Texas Health Sciences Center Houston, Houston, Texas, USA
| | - Samuel A. Shelburne
- Department of Infectious Disease, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
7
|
Hayama B, Harada S, Suzuki M, Doi Y, Nomura Y, Aoki K, Takehana K, Akatsuchi T, Enokida T, Takeda K, Seto A, Mitani H, Ohkushi D. Outbreak of Streptococcus pyogenes emm89 ST646 in a head and neck surgical oncology ward. Microbiol Spectr 2024; 12:e0426023. [PMID: 38587390 PMCID: PMC11064547 DOI: 10.1128/spectrum.04260-23] [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: 12/20/2023] [Accepted: 03/18/2024] [Indexed: 04/09/2024] Open
Abstract
Streptococcus pyogenes causes a variety of human infections, and hospital outbreaks with this pathogen have also been reported. The purpose of this study is to describe the clinical characteristics of an outbreak of S. pyogenes involving 15 patients and four healthcare workers (HCWs), as well as the molecular characteristics of the causative isolates. The course and response to the outbreak were reviewed, and information on the characteristics of the patients was extracted retrospectively from the medical records. Whole-genome sequencing of the 16 causative isolates (14 from patients and two from HCWs) was also performed. All 15 patients were postoperative of head and neck cancer with tracheotomy, and 12 had invasive infections, primarily surgical site infections, all of which resolved without causing serious illness. All but the first case was detected more than 7 days after admission. S. pyogenes was detected in two patients after empiric antimicrobial administration was performed on all inpatients and HCWs, and the outbreak was finally contained in approximately 2 months. All isolates detected in patients and HCWs belonged to emm89/clade 3, a hypervirulent clone that has emerged worldwide and was classified as sequence type 646. These isolates had single nucleotide polymorphism (SNP) differences of zero to one, indicating clonal transmission. This study demonstrated an outbreak of S. pyogenes emm89/clade 3 in a ward of patients with head and neck cancer. The global emergence of hypervirulent isolates may increase the risk of outbreaks among high-risk patients. IMPORTANCE This study describes an outbreak of Streptococcus pyogenes that occurred in a ward caring for patients with head and neck cancer and tracheostomies. Many cases of invasive infections occurred in a short period, and extensive empiric antimicrobial administration on patients and healthcare workers was performed to control the outbreak. Whole-genome sequencing analysis of the causative strains confirmed that it was a monoclonal transmission of strains belonging to emm89/clade 3. The epidemiology and clinical characteristics of S. pyogenes infections have changed with the replacement of the prevalent clones worldwide. In the 1980s, there was a reemergence of S. pyogenes infections in high-income countries due to the spread of hypervirulent emm1 strains. emm89/clade 3 has recently been spreading worldwide and shares common features with emm1, including increased production of two toxins, NADase, and streptolysin O. The outbreak reported here may reflect the high spreading potential and virulence of emm89/clade 3.
Collapse
Affiliation(s)
- Brian Hayama
- Department of Infectious Diseases, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Infection Prevention, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Sohei Harada
- Department of Infectious Diseases, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, Tokyo, Japan
| | - Masahiro Suzuki
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yohei Doi
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yusuke Nomura
- Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan
| | - Kotaro Aoki
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, Tokyo, Japan
| | - Kazumi Takehana
- Clinical Laboratories, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomomi Akatsuchi
- Department of Infection Prevention, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Taisuke Enokida
- Department of Infectious Diseases, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koichi Takeda
- Department of Infectious Diseases, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akira Seto
- Department of Head and Neck Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroki Mitani
- Department of Head and Neck Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Daisuke Ohkushi
- Department of Infectious Diseases, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
8
|
Odo CM, Vega LA, Mukherjee P, DebRoy S, Flores AR, Shelburne SA. Emergent emm4 group A Streptococcus evidences a survival strategy during interaction with immune effector cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588776. [PMID: 38645060 PMCID: PMC11030381 DOI: 10.1101/2024.04.09.588776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The major gram-positive pathogen group A Streptococcus (GAS) is a model organism for studying microbial epidemics as it causes waves of infections. Since 1980, several GAS epidemics have been ascribed to the emergence of clones producing increased amounts of key virulence factors such as streptolysin O (SLO). Herein, we sought to identify mechanisms underlying our recently identified temporal clonal emergence amongst emm4 GAS, given that emergent strains did not produce augmented levels of virulence factors relative to historic isolates. Through the creation and analysis of isoallelic strains, we determined that a conserved mutation in a previously undescribed gene encoding a putative carbonic anhydrase was responsible for the defective in vitro growth observed in the emergent strains. We also identified that the emergent strains survived better inside macrophages and killed macrophages at lower rates relative to the historic strains. Via creation of isogenic mutant strains, we linked the emergent strain "survival" phenotype to the downregulation of the SLO encoding gene and upregulation of the msrAB operon which encodes proteins involved in defense against extracellular oxidative stress. Our findings are in accord with recent surveillance studies which found high ratio of mucosal (i.e., pharyngeal) relative to invasive infections amongst emm4 GAS. Inasmuch as ever-increasing virulence is unlikely to be evolutionary advantageous for a microbial pathogen, our data furthers understanding of the well described oscillating patterns of virulent GAS infections by demonstrating mechanisms by which emergent strains adapt a "survival" strategy to outcompete previously circulating isolates.
Collapse
|
9
|
Takahashi R, J-Khemlani AH, Loh JMS, Radcliff FJ, Proft T, Tsai CJY. Different Group A Streptococcus pili lead to varying proinflammatory cytokine responses and virulence. Immunol Cell Biol 2024; 102:21-33. [PMID: 37795567 DOI: 10.1111/imcb.12692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
The human pathogen Streptococcus pyogenes, or Group A Streptococcus (GAS), is associated with a variety of diseases ranging from mild skin and soft tissue infections to invasive diseases and immune sequelae such as rheumatic heart disease. We have recently reported that one of the virulence factors of this pathogen, the pilus, has inflammatory properties and strongly stimulates the innate immune system. Here we used a range of nonpathogenic Lactococcus lactis gain-of-function mutants, each expressing one of the major pilus types of GAS, to compare the immune responses generated by various types of fully assembled pili. In vitro assays indicated variability in the inflammatory response induced by different pili, with the fibronectin-binding, collagen-binding, T antigen (FCT)-1-type pilus from GAS serotype M6/T6 inducing significantly stronger cytokine secretion than other pili. Furthermore, we established that the same trend of pili-mediated immune response could be modeled in Galleria mellonella larvae, which possess a similar innate immune system to vertebrates. Counterintuitively, across the panel of pili types examined in this study, we observed a negative correlation between the intensity of the immune response demonstrated in our experiments and the disease severity observed clinically in the GAS strains associated with each pilus type. This observation suggests that pili-mediated inflammation is more likely to promote bacterial clearance instead of causing disruptive damages that intensify pathogenesis. This also indicates that pili may not be the main contributor to the inflammatory symptoms seen in GAS diseases. Rather, the immune-potentiating properties of the pilus components could potentially be exploited as a vaccine adjuvant.
Collapse
Affiliation(s)
- Risa Takahashi
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Adrina Hema J-Khemlani
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Jacelyn Mei San Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Fiona Jane Radcliff
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Catherine Jia-Yun Tsai
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Moriel DG, Piccioli D, Raso MM, Pizza M. The overlooked bacterial pandemic. Semin Immunopathol 2024; 45:481-491. [PMID: 38078911 PMCID: PMC11136754 DOI: 10.1007/s00281-023-00997-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/28/2023] [Indexed: 05/30/2024]
Abstract
The COVID-19 pandemic had a significant economic and health impact worldwide. It also reinforced the misperception that only viruses can pose a threat to human existence, overlooking that bacteria (e.g., plague and cholera) have severely haunted and shaped the course of human civilization. While the world is preparing for the next viral pandemic, it is again overlooking a silent one: antimicrobial resistance (AMR). This review proposes to show the impact of bacterial infections on civilization to remind the pandemic potential. The work will also discuss a few examples of how bacteria can mutate risking global spread and devastating outcomes, the effect on the global burden, and the prophylactic and therapeutic measures. Indeed, AMR is dramatically increasing and if the trend is not reversed, it has the potential to quickly turn into the most important health problem worldwide.
Collapse
Affiliation(s)
- Danilo Gomes Moriel
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy
| | - Diego Piccioli
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy
| | | | | |
Collapse
|
11
|
Ho EC, Cataldi JR, Silveira LJ, Birkholz M, Loi MM, Osborne CM, Dominguez SR. Outbreak of Invasive Group A Streptococcus in Children-Colorado, October 2022-April 2023. J Pediatric Infect Dis Soc 2023; 12:540-548. [PMID: 37792995 DOI: 10.1093/jpids/piad080] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND In the fall of 2022, we observed a sharp rise in pediatric Invasive Group A Streptococcus (iGAS) hospitalizations in Colorado. We compared the epidemiology, clinical features, and patient outcomes in this outbreak to prior years. METHODS Between October 2022 and April 2023, we prospectively identified and reviewed iGAS cases in hospitalized pediatric patients at Children's Hospital Colorado. Using laboratory specimen records, we also retrospectively compared the number of patients with sterile site GAS-positive cultures across three time periods: pre-COVID-19 (January 2015-March 2020), height of COVID-19 pandemic (April 2020-September 2022), and outbreak (October 2022-April 2023). RESULTS Among 96 prospectively identified iGAS cases, median age was 5.7 years old; 66% were male, 70% previously healthy, 39% required critical care, and four patients died. Almost 60% had associated respiratory viral symptoms, 10% had toxic shock syndrome, and 4% had necrotizing fasciitis. Leukopenia, bandemia, and higher C-reactive protein values were laboratory findings associated with need for critical care. There were significantly more cases during the outbreak (9.9/month outbreak vs 3.9/month pre-pandemic vs 1.3/month pandemic), including more cases with pneumonia (28% outbreak vs 15% pre-pandemic vs 0% pandemic) and multifocal disease (17% outbreak vs 3% pre-pandemic vs 0% pandemic), P < .001 for all. CONCLUSIONS Outbreak case numbers were almost triple the pre-pandemic baseline. The high percentage of cases with associated viral symptoms suggests a link to coinciding surges in respiratory viruses during this time. Invasive GAS can be severe and evolve rapidly; clinical and laboratory features may help in earlier identification of critically ill children.
Collapse
Affiliation(s)
- Erin C Ho
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
- Section of Infectious Diseases, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Jessica R Cataldi
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
- Section of Infectious Diseases, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Lori J Silveira
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Meghan Birkholz
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Michele M Loi
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
- Section of Critical Care Medicine, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Christina M Osborne
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA and
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Samuel R Dominguez
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
- Section of Infectious Diseases, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
12
|
Nookala S, Mukundan S, Grove B, Combs C. Concurrent Brain Subregion Microgliosis in an HLA-II Mouse Model of Group A Streptococcal Skin Infection. Microorganisms 2023; 11:2356. [PMID: 37764200 PMCID: PMC10538044 DOI: 10.3390/microorganisms11092356] [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: 07/17/2023] [Revised: 08/28/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
The broad range of clinical manifestations and life-threatening infections caused by the Gram-positive bacterium, Streptococcus pyogenes or Group A Streptococcus (GAS), remains a significant concern to public health, with a subset of individuals developing neurological complications. Here, we examined the concurrent neuroimmune effects of subcutaneous GAS infections in an HLA-Class II (HLA) transgenic mouse model of subcutaneous GAS infection. To investigate changes in the skin-brain axis, HLA-DQ8 (DQA1*0301/DQB1*0302) mice (DQ8) were randomly divided into three groups: uninfected controls (No Inf), GAS infected and untreated (No Tx), and GAS infected with a resolution by clindamycin (CLN) treatment (CLN Tx) (10 mg/kg/5 days) and were monitored for 16 days post-infection. While the skin GAS burden was significantly reduced by CLN, the cortical and hippocampal GAS burden in the male DQ8 mice was not significantly reduced with CLN. Immunoreactivity to anti-GAS antibody revealed the presence of GAS bacteria in the vicinity of the neuronal nucleus in the neocortex of both No Tx and CLN Tx male DQ8 mice. GAS infection-mediated cortical cytokine changes were modest; however, compared to No Inf or No Tx groups, a significant increase in IL-2, IL-13, IL-22, and IL-10 levels was observed in CLN Tx females despite the lack of GAS burden. Western blot analysis of cortical and hippocampal homogenates showed significantly higher ionized calcium-binding adaptor-1 (Iba-1, microglia marker) protein levels in No Tx females and males and CLN Tx males compared to the No Inf group. Immunohistochemical analysis showed that Iba-1 immunoreactivity in the hippocampal CA3 and CA1 subregions was significantly higher in the CLN Tx males compared to the No Tx group. Our data support the possibility that the subcutaneous GAS infection communicates to the brain and is characterized by intraneuronal GAS sequestration, brain cytokine changes, Iba-1 protein levels, and concurrent CA3 and CA1 subregion-specific microgliosis, even without bacteremia.
Collapse
Affiliation(s)
- Suba Nookala
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.M.); (B.G.); (C.C.)
| | | | | | | |
Collapse
|
13
|
Davies MR, Keller N, Brouwer S, Jespersen MG, Cork AJ, Hayes AJ, Pitt ME, De Oliveira DMP, Harbison-Price N, Bertolla OM, Mediati DG, Curren BF, Taiaroa G, Lacey JA, Smith HV, Fang NX, Coin LJM, Stevens K, Tong SYC, Sanderson-Smith M, Tree JJ, Irwin AD, Grimwood K, Howden BP, Jennison AV, Walker MJ. Detection of Streptococcus pyogenes M1 UK in Australia and characterization of the mutation driving enhanced expression of superantigen SpeA. Nat Commun 2023; 14:1051. [PMID: 36828918 PMCID: PMC9951164 DOI: 10.1038/s41467-023-36717-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
A new variant of Streptococcus pyogenes serotype M1 (designated 'M1UK') has been reported in the United Kingdom, linked with seasonal scarlet fever surges, marked increase in invasive infections, and exhibiting enhanced expression of the superantigen SpeA. The progenitor S. pyogenes 'M1global' and M1UK clones can be differentiated by 27 SNPs and 4 indels, yet the mechanism for speA upregulation is unknown. Here we investigate the previously unappreciated expansion of M1UK in Australia, now isolated from the majority of serious infections caused by serotype M1 S. pyogenes. M1UK sub-lineages circulating in Australia also contain a novel toxin repertoire associated with epidemic scarlet fever causing S. pyogenes in Asia. A single SNP in the 5' transcriptional leader sequence of the transfer-messenger RNA gene ssrA drives enhanced SpeA superantigen expression as a result of ssrA terminator read-through in the M1UK lineage. This represents a previously unappreciated mechanism of toxin expression and urges enhanced international surveillance.
Collapse
Affiliation(s)
- Mark R Davies
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | - Nadia Keller
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Stephan Brouwer
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Magnus G Jespersen
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Amanda J Cork
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew J Hayes
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Miranda E Pitt
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David M P De Oliveira
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nichaela Harbison-Price
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Olivia M Bertolla
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel G Mediati
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Bodie F Curren
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - George Taiaroa
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jake A Lacey
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Helen V Smith
- Public Health Microbiology, Queensland Health Forensic and Scientific Services, Queensland Health, Coopers Plains, QLD, Australia
| | - Ning-Xia Fang
- Public Health Microbiology, Queensland Health Forensic and Scientific Services, Queensland Health, Coopers Plains, QLD, Australia
| | - Lachlan J M Coin
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kerrie Stevens
- Microbiological Diagnostic Unit Public Health Laboratory, The Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Steven Y C Tong
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Victorian Infectious Diseases Service, The Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Martina Sanderson-Smith
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Jai J Tree
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Adam D Irwin
- University of Queensland Centre for Clinical Research, Brisbane, QLD, Australia.,Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Keith Grimwood
- School of Medicine and Dentistry and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Departments of Infectious Diseases and Paediatrics, Gold Coast Health, Gold Coast, QLD, Australia
| | - Benjamin P Howden
- Microbiological Diagnostic Unit Public Health Laboratory, The Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Amy V Jennison
- Public Health Microbiology, Queensland Health Forensic and Scientific Services, Queensland Health, Coopers Plains, QLD, Australia
| | - Mark J Walker
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
14
|
Anand A, Sharma A, Ravins M, Johri AK, Tirosh B, Hanski E. PERK Pathway Inhibitors Cure Group A Streptococcal Necrotizing Fasciitis in a Murine Model. Bio Protoc 2022; 12:e4589. [PMID: 36618091 PMCID: PMC9797358 DOI: 10.21769/bioprotoc.4589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/05/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Group A streptococcus (GAS) is a Gram-positive human pathogen that causes invasive infections with mild to life-threatening severity, like toxic shock syndrome, rheumatic heart disease, and necrotizing fasciitis (NF). NF is characterized by a clinical presentation of widespread tissue destruction due to the rapid spread of GAS infection into fascial planes. Despite quick medical interventions, mortality from NF is high. The early onset of the disease is difficult to diagnose because of non-specific clinical symptoms. Moreover, the unavailability of an effective vaccine against GAS warrants a genuine need for alternative treatments against GAS NF. One endoplasmic reticulum stress signaling pathway (PERK pathway) gets triggered in the host upon GAS infection. Bacteria utilize asparagine release as an output of this pathway for its pathogenesis. We reported that the combination of sub-cutaneous (SC) and intraperitoneal (IP) administration of PERK pathway inhibitors (GSK2656157 and ISRIB) cures local as well as systemic GAS infection in a NF murine model, by reducing asparagine release at the infection site. This protocol's methodology is detailed below. This protocol was validated in: Sci Transl Med (2021), DOI: 10.1126/scitranslmed.abd7465.
Collapse
Affiliation(s)
- Aparna Anand
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Abhinay Sharma
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Miriam Ravins
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India
| | - Boaz Tirosh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Emanuel Hanski
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
,
*For correspondence:
| |
Collapse
|
15
|
Bergmann R, Gulotta G, Andreoni F, Sumitomo T, Kawabata S, Zinkernagel AS, Chhatwal GS, Nizet V, Rohde M, Uchiyama S. The group A Streptococcus interleukin-8 protease SpyCEP promotes bacterial intracellular survival by evasion of autophagy. INFECTIOUS MICROBES & DISEASES 2022; 4:116-123. [PMID: 37333426 PMCID: PMC10275413 DOI: 10.1097/im9.0000000000000098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Autophagy serves an innate immune function in defending the host against invading bacteria, including group A Streptococcus (GAS). Autophagy is regulated by numerous host proteins, including the endogenous negative regulator calpain, a cytosolic protease. Globally disseminated serotype M1T1 GAS strains associated with high invasive disease potential express numerous virulence factors and resist autophagic clearance. Upon in vitro infection of human epithelial cell lines with representative wild-type GAS M1T1 strain 5448 (M1.5448), we observed increased calpain activation linked to a specific GAS virulence factor, the IL-8 protease SpyCEP. Calpain activation inhibited autophagy and decreased capture of cytosolic GAS in autophagosomes. In contrast, the serotype M6 GAS strain JRS4 (M6.JRS4), which is highly susceptible to host autophagy-mediated killing, expresses low levels of SpyCEP and does not activate calpain. Overexpression of SpyCEP in M6.JRS4 stimulated calpain activation, inhibited autophagy and significantly decreased bacterial capture in autophagosomes. These paired loss- and gain-of-function studies reveal a novel role for the bacterial protease SpyCEP in enabling GAS M1 evasion of autophagy and host innate immune clearance.
Collapse
Affiliation(s)
- René Bergmann
- Central Unit for Microscopy (ZEIM), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Giuseppe Gulotta
- Central Unit for Microscopy (ZEIM), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Federica Andreoni
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Tomoko Sumitomo
- Department of Oral and Molecular Microbiology, 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
| | - Annelies S. Zinkernagel
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Gursharan S. Chhatwal
- Central Unit for Microscopy (ZEIM), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Manfred Rohde
- Central Unit for Microscopy (ZEIM), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Satoshi Uchiyama
- Central Unit for Microscopy (ZEIM), Helmholtz Centre for Infection Research, Braunschweig, Germany
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Rom JS, Le Breton Y, Islam E, Belew AT, El-Sayed NM, McIver KS. Loss of rpoE Encoding the δ-Factor of RNA Polymerase Impacts Pathophysiology of the Streptococcus pyogenes M1T1 Strain 5448. Microorganisms 2022; 10:microorganisms10081686. [PMID: 36014103 PMCID: PMC9412562 DOI: 10.3390/microorganisms10081686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/08/2022] [Accepted: 08/19/2022] [Indexed: 12/01/2022] Open
Abstract
Streptococcus pyogenes, also known as the Group A Streptococcus (GAS), is a Gram-positive bacterial pathogen of major clinical significance. Despite remaining relatively susceptible to conventional antimicrobial therapeutics, GAS still causes millions of infections and hundreds of thousands of deaths each year worldwide. Thus, a need for prophylactic and therapeutic interventions for GAS is in great demand. In this study, we investigated the importance of the gene encoding the delta (δ) subunit of the GAS RNA polymerase, rpoE, for its impact on virulence during skin and soft-tissue infection. A defined 5448 mutant with an insertionally-inactivated rpoE gene was defective for survival in whole human blood and was attenuated for both disseminated lethality and lesion size upon mono-culture infection in mouse soft tissue. Furthermore, the mutant had reduced competitive fitness when co-infected with wild type (WT) 5448 in the mouse model. We were unable to attribute this attenuation to any observable growth defect, although colony size and the ability to grow at higher temperatures were both affected when grown with nutrient-rich THY media. RNA-seq of GAS grown in THY to late log phase found that mutation of rpoE significantly impacted (>2-fold) the expression of 429 total genes (205 upregulated, 224 downregulated), including multiple virulence and “housekeeping” genes. The arc operon encoding the arginine deiminase (ADI) pathway was the most upregulated in the rpoE mutant and this could be confirmed phenotypically. Taken together, these findings demonstrate that the delta (δ) subunit of RNA polymerase is vital in GAS gene expression and virulence.
Collapse
|
17
|
De Oliveira DMP, Keller B, Hayes AJ, Ong CLY, Harbison-Price N, El-Deeb IM, Li G, Keller N, Bohlmann L, Brouwer S, Turner AG, Cork AJ, Jones TR, Paterson DL, McEwan AG, Davies MR, McDevitt CA, von Itzstein M, Walker MJ. Neurodegenerative Disease Treatment Drug PBT2 Breaks Intrinsic Polymyxin Resistance in Gram-Positive Bacteria. Antibiotics (Basel) 2022; 11:antibiotics11040449. [PMID: 35453201 PMCID: PMC9027797 DOI: 10.3390/antibiotics11040449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 12/03/2022] Open
Abstract
Gram-positive bacteria do not produce lipopolysaccharide as a cell wall component. As such, the polymyxin class of antibiotics, which exert bactericidal activity against Gram-negative pathogens, are ineffective against Gram-positive bacteria. The safe-for-human-use hydroxyquinoline analog ionophore PBT2 has been previously shown to break polymyxin resistance in Gram-negative bacteria, independent of the lipopolysaccharide modification pathways that confer polymyxin resistance. Here, in combination with zinc, PBT2 was shown to break intrinsic polymyxin resistance in Streptococcus pyogenes (Group A Streptococcus; GAS), Staphylococcus aureus (including methicillin-resistant S. aureus), and vancomycin-resistant Enterococcus faecium. Using the globally disseminated M1T1 GAS strain 5448 as a proof of principle model, colistin in the presence of PBT2 + zinc was shown to be bactericidal in activity. Any resistance that did arise imposed a substantial fitness cost. PBT2 + zinc dysregulated GAS metal ion homeostasis, notably decreasing the cellular manganese content. Using a murine model of wound infection, PBT2 in combination with zinc and colistin proved an efficacious treatment against streptococcal skin infection. These findings provide a foundation from which to investigate the utility of PBT2 and next-generation polymyxin antibiotics for the treatment of Gram-positive bacterial infections.
Collapse
Affiliation(s)
- David M. P. De Oliveira
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Bernhard Keller
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Andrew J. Hayes
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; (A.J.H.); (M.R.D.); (C.A.M.)
| | - Cheryl-Lynn Y. Ong
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Nichaela Harbison-Price
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Ibrahim M. El-Deeb
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia; (I.M.E.-D.); (M.v.I.)
| | - Gen Li
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Nadia Keller
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Lisa Bohlmann
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Stephan Brouwer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Andrew G. Turner
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Amanda J. Cork
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Thomas R. Jones
- School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - David L. Paterson
- Australian Infectious Diseases Research Centre, UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4006, Australia;
| | - Alastair G. McEwan
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
| | - Mark R. Davies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; (A.J.H.); (M.R.D.); (C.A.M.)
| | - Christopher A. McDevitt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; (A.J.H.); (M.R.D.); (C.A.M.)
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia; (I.M.E.-D.); (M.v.I.)
| | - Mark J. Walker
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (D.M.P.D.O.); (B.K.); (C.-L.Y.O.); (N.H.-P.); (G.L.); (N.K.); (L.B.); (S.B.); (A.G.T.); (A.J.C.); (A.G.M.)
- Correspondence: ; Tel.: +61-7-33461623
| |
Collapse
|
18
|
Indraratna AD, Everest-Dass A, Skropeta D, Sanderson-Smith M. OUP accepted manuscript. FEMS Microbiol Rev 2022; 46:6519265. [PMID: 35104861 PMCID: PMC9075583 DOI: 10.1093/femsre/fuac001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/29/2021] [Accepted: 01/25/2022] [Indexed: 11/12/2022] Open
Abstract
Host carbohydrates, or glycans, have been implicated in the pathogenesis of many bacterial infections. Group A Streptococcus (GAS) is a Gram-positive bacterium that readily colonises the skin and oropharynx, and is a significant cause of mortality in humans. While the glycointeractions orchestrated by many other pathogens are increasingly well-described, the understanding of the role of human glycans in GAS disease remains incomplete. Although basic investigation into the mechanisms of GAS disease is ongoing, several glycointeractions have been identified and are examined herein. The majority of research in this context has focussed on bacterial adherence, however, glycointeractions have also been implicated in carbohydrate metabolism; evasion of host immunity; biofilm adaptations; and toxin-mediated haemolysis. The involvement of human glycans in these diverse avenues of pathogenesis highlights the clinical value of understanding glycointeractions in combatting GAS disease.
Collapse
Affiliation(s)
- Anuk D Indraratna
- Illawarra Health and Medical Research Institute, Northfields Ave, Keiraville New South Wales 2522, Australia
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia
| | - Arun Everest-Dass
- Institute for Glycomics, Griffith University, Gold Coast Campus, Parklands Drive, Southport, Queensland, 4215, Australia
| | - Danielle Skropeta
- Illawarra Health and Medical Research Institute, Northfields Ave, Keiraville New South Wales 2522, Australia
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia
| | - Martina Sanderson-Smith
- Corresponding author: Illawarra Health and Medical Research Institute, Bld 32, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia. Tel: +61 2 42981935; E-mail:
| |
Collapse
|
19
|
Abstract
Streptococcus pyogenes (group A Streptococcus) is a globally disseminated and human-adapted bacterial pathogen that causes a wide range of infections, including scarlet fever. Scarlet fever is a toxin-mediated disease characterized by the formation of an erythematous, sandpaper-like rash that typically occurs in children aged 5 to 15. This infectious disease is caused by toxins called superantigens, a family of highly potent immunomodulators. Although scarlet fever had largely declined in both prevalence and severity since the late 19th century, outbreaks have now reemerged in multiple geographical regions over the past decade. Here, we review recent findings that address the role of superantigens in promoting a fitness advantage for S. pyogenes within human populations and discuss how superantigens may be suitable targets for vaccination strategies.
Collapse
Affiliation(s)
- Jacklyn R. Hurst
- Department of Microbiology and Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Stephan Brouwer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Mark J. Walker
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
- * E-mail: (MJW); (JKM)
| | - John K. McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- * E-mail: (MJW); (JKM)
| |
Collapse
|
20
|
Host-to-Host Group A Streptococcus Transmission Causes Infection of the Lamina Propria but not Epithelium of the Upper Respiratory Tract in MyD88-Deficient Mice. Infect Immun 2021; 90:e0042321. [PMID: 34662211 DOI: 10.1128/iai.00423-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To understand protective immune responses against the onset of Group A Streptococcus respiratory infection, we investigated whether MyD88 KO mice were susceptible to acute infection through transmission. After commingling with mice that had intranasal GAS inoculation, MyD88-/- recipient mice had increased GAS loads in the nasal cavity and throat that reached a mean throat colonization of 6.3 x 106 cfu/swab and mean GAS load of 5.2 x 108 cfu in the nasal cavity on day 7. Beyond day 7, MyD88-/- recipient mice became moribund, with mean 1.6 x 107 cfu/swab and 2.5 x 109 cfu GAS in the throat and nasal cavity, respectively. Systemic GAS infection occurred a couple of days after the upper respiratory infection. GAS infects the lip, gingival sulcus of the incisor teeth, the lamina propria of the turbinate but not the nasal cavity and nasopharyngeal tract epithelia, and C57BL/6J recipient mice had no or low levels of GAS in the nasal cavity and throat. Direct nasal GAS inoculation of MyD88-/- mice caused GAS infection mainly in the lamina propria of the turbinate. In contrast, C57BL/6J mice with GAS inoculation had GAS bacteria in the nasal cavity but not in the lamina propria of the turbinates. Thus, MyD88-/- mice are highly susceptible to acute and lethal GAS infection through transmission, and MyD88 signaling is critical for protection of the respiratory tract lamina propria but not nasal and nasopharyngeal epithelia against GAS infection.
Collapse
|
21
|
Richter J, Monteleone MM, Cork AJ, Barnett TC, Nizet V, Brouwer S, Schroder K, Walker MJ. Streptolysins are the primary inflammasome activators in macrophages during Streptococcus pyogenes infection. Immunol Cell Biol 2021; 99:1040-1052. [PMID: 34462965 DOI: 10.1111/imcb.12499] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/09/2021] [Accepted: 08/28/2021] [Indexed: 12/30/2022]
Abstract
Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes an array of infectious diseases in humans. Accumulating clinical evidence suggests that proinflammatory interleukin (IL)-1β signaling plays an important role in GAS disease progression. The host regulates the production and secretion of IL-1β via the cytosolic inflammasome pathway. Activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome complex requires two signals: a priming signal that stimulates increased transcription of genes encoding the components of the inflammasome pathway, and an activating signal that induces assembly of the inflammasome complex. Here we show that GAS-derived lipoteichoic acid can provide a priming signal for NLRP3 inflammasome activation. As only few GAS-derived proteins have been associated with inflammasome-dependent IL-1β signaling, we investigated novel candidates that might play a role in activating the inflammasome pathway by infecting mouse bone marrow-derived macrophages and human THP-1 macrophage-like cells with a panel of isogenic GAS mutant strains. We found that the cytolysins streptolysin O (SLO) and streptolysin S are the main drivers of IL-1β release in proliferating logarithmic phase GAS. Using a mutant form of recombinant SLO, we confirmed that bacterial pore formation on host cell membranes is a key mechanism required for inflammasome activation. Our results suggest that streptolysins are major determinants of GAS-induced inflammation and present an attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Johanna Richter
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mercedes M Monteleone
- Australian Infectious Diseases Research Centre, Institute for Molecular Bioscience and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia, QLD, Australia
| | - Amanda J Cork
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Timothy C Barnett
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Victor Nizet
- Department of Pediatrics, University of California at San Diego School of Medicine, La Jolla, CA, USA.,Skaggs School of Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA, USA
| | - Stephan Brouwer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Kate Schroder
- Australian Infectious Diseases Research Centre, Institute for Molecular Bioscience and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia, QLD, Australia
| | - Mark J Walker
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
22
|
Anand A, Sharma A, Ravins M, Biswas D, Ambalavanan P, Lim KXZ, Tan RYM, Johri AK, Tirosh B, Hanski E. Unfolded protein response inhibitors cure group A streptococcal necrotizing fasciitis by modulating host asparagine. Sci Transl Med 2021; 13:13/605/eabd7465. [PMID: 34349034 DOI: 10.1126/scitranslmed.abd7465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/24/2021] [Accepted: 06/25/2021] [Indexed: 11/02/2022]
Abstract
Group A streptococcus (GAS) is among the top 10 causes of mortality from an infectious disease, producing mild to invasive life-threatening manifestations. Necrotizing fasciitis (NF) is characterized by a rapid GAS spread into fascial planes followed by extensive tissue destruction. Despite prompt treatments of antibiotic administration and tissue debridement, mortality from NF is still high. Moreover, there is no effective vaccine against GAS, and early diagnosis of NF is problematic because its clinical presentations are not specific. Thus, there is a genuine need for effective treatments against GAS NF. Previously, we reported that GAS induces endoplasmic reticulum (ER) stress to gain asparagine from the host. Here, we demonstrate that GAS-mediated asparagine induction and release occur through the PERK-eIF2α-ATF4 branch of the unfolded protein response. Inhibitors of PERK or integrated stress response (ISR) blocked the formation and release of asparagine by infected mammalian cells, and exogenously added asparagine overcame this inhibition. Moreover, in a murine model of NF, we show that the inhibitors minimized mortality when mice were challenged with a lethal dose of GAS and reduced bacterial counts and lesion size when mice were challenged with a sublethal dose. Immunohistopathology studies demonstrated that PERK/ISR inhibitors protected mice by enabling neutrophil infiltration into GAS-infected fascia and reducing the pro-inflammatory response that causes tissue damage. Inhibitor treatment was also effective in mice when started at 12 hours after infection. We conclude that host metabolic alteration induced by PERK or ISR inhibitors is a promising therapeutic strategy to treat highly invasive GAS infections.
Collapse
Affiliation(s)
- Aparna Anand
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Abhinay Sharma
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Miriam Ravins
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Debabrata Biswas
- Singapore-HUJ Alliance for Research and Enterprise, MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 117576, Singapore.,Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| | - Poornima Ambalavanan
- Singapore-HUJ Alliance for Research and Enterprise, MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 117576, Singapore.,Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| | - Kimberly Xuan Zhen Lim
- Singapore-HUJ Alliance for Research and Enterprise, MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 117576, Singapore.,Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| | - Rachel Ying Min Tan
- Singapore-HUJ Alliance for Research and Enterprise, MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 117576, Singapore.,Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India
| | - Boaz Tirosh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel.
| | - Emanuel Hanski
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel. .,Singapore-HUJ Alliance for Research and Enterprise, MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 117576, Singapore.,Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
23
|
Wright CM, Langworthy K, Manning L. The Australian burden of invasive group A streptococcal disease: a narrative review. Intern Med J 2021; 51:835-844. [PMID: 32372512 DOI: 10.1111/imj.14885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022]
Abstract
The Australian and New Zealand governments have allocated significant funding to advance efforts towards a group A Streptococcus (Strep A) vaccine. The argument for Strep A vaccine development has to date focussed on prevention of non-invasive disease (e.g. pharyngitis) and immune-mediated complications (especially rheumatic heart disease). Because of the poorer prognosis and theoretically more precisely known burden of invasive, compared to non-invasive disease, exploration of the burden of invasive Strep A disease could lend further support to the vaccine business case. This narrative review critically assesses the Australian incidence of invasive Strep A disease. Case notification data were first assessed through government sources, expressing annual incidence as cases per 100 000 population. Published literature accessed through PubMed and MEDLINE was assessed to March 2020. Where estimates could be updated by replicating reported methods with publicly available data, this was performed. Invasive Strep A disease is currently notifiable in Queensland and the Northern Territory only. The magnitude, degree of certainty and recency of estimates vary by state/territory and between sub-populations, including higher incidence among Indigenous Australians compared to non-Indigenous Australians. According to inpatient records from 2017 to 2018, the Australian incidence of invasive Strep A disease was 8.3 per 100 000. However, this is likely to be an underestimate. Preventing invasive Strep A disease is an important use for a Strep A vaccine. This narrative review highlights deficiencies in our current understanding of the Australian disease burden. These difficulties would be overcome by nationally consistent mandatory case reporting.
Collapse
Affiliation(s)
- Cameron M Wright
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia.,Health Economics and Data Analytics, School of Public Health, Faculty of Health Sciences, Curtin University, Perth, Western Australia, Australia.,School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kristyn Langworthy
- Department of Infectious Diseases, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Laurens Manning
- Department of Infectious Diseases, Fiona Stanley Hospital, Perth, Western Australia, Australia.,Faculty of Health and Medical Sciences, Harry Perkins Research Institute, Fiona Stanley Hospital, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
24
|
Revisiting the inoculum effect for Streptococcus pyogenes with a hollow fibre infection model. Eur J Clin Microbiol Infect Dis 2021; 40:2137-2144. [PMID: 33948751 DOI: 10.1007/s10096-021-04262-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Severe, invasive Streptococcus pyogenes (Strep A) infections result in greater than 500,000 deaths annually. First line treatment for such infections is benzylpenicillin, often with the addition of clindamycin, but treatment failure can occur with this regimen. This failure has been partially attributed to the inoculum effect, which presents as reduced antibiotic susceptibility during high bacterial density and plateau-phase growth. Hollow fibre infection models (HFIM) have been proposed as an in vitro alternative to in vivo research to study these effects. To re-evaluate the inoculum effect for benzylpenicillin, clindamycin, linezolid, and trimethoprim-sulfamethoxazole using a Strep A HFIM. Differential antibiotic susceptibility of Strep A was measured in a HFIM starting from low- and high-density inocula with an average difference in bacterial concentration of 56-fold. Dynamic antibiotic concentrations were delivered over 48 h to simulate in vivo human pharmacokinetics in an in vitro model. Differences in antibiotic susceptibility were measured by plate count of colony-forming units over time. Inoculum effects were seen in benzylpenicillin and linezolid at 24 h, and benzylpenicillin, linezolid, and clindamycin at 48 h. The effect size was greatest for continuously infused benzylpenicillin at 48 h with a log10-fold difference of 4.02 between groups. No inoculum effect was seen in trimethoprim-sulfamethoxazole, with a maximal log10-fold difference of 0.40. Inoculum effects were seen using benzylpenicillin, linezolid, and clindamycin, which may predict reduced clinical efficacy following treatment delay. The model has proven robust and largely in agreeance with published data, recommending it for further Strep A study.
Collapse
|
25
|
Williams JG, Ly D, Geraghty NJ, McArthur JD, Vyas HKN, Gorman J, Tsatsaronis JA, Sluyter R, Sanderson-Smith ML. Streptococcus pyogenes M1T1 Variants Induce an Inflammatory Neutrophil Phenotype Including Activation of Inflammatory Caspases. Front Cell Infect Microbiol 2021; 10:596023. [PMID: 33585270 PMCID: PMC7876443 DOI: 10.3389/fcimb.2020.596023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Invasive infections due to group A Streptococcus (GAS) advance rapidly causing tissue degradation and unregulated inflammation. Neutrophils are the primary immune cells that respond to GAS. The neutrophil response to GAS was characterised in response to two M1T1 isolates; 5448 and animal passaged variant 5448AP. Co-incubation of neutrophils with 5448AP resulted in proliferation of GAS and lowered the production of reactive oxygen species when compared with 5448. Infection with both strains invoked neutrophil death, however apoptosis was reduced in response to 5448AP. Both strains induced neutrophil caspase-1 and caspase-4 expression in vitro, with inflammatory caspase activation detected in vitro and in vivo. GAS infections involving strains such as 5448AP that promote an inflammatory neutrophil phenotype may contribute to increased inflammation yet ineffective bacterial eradication, contributing to the severity of invasive GAS infections.
Collapse
Affiliation(s)
- Jonathan G. Williams
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Diane Ly
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Nicholas J. Geraghty
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Jason D. McArthur
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Heema K. N. Vyas
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Jody Gorman
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - James A. Tsatsaronis
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Ronald Sluyter
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Martina L. Sanderson-Smith
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
26
|
Sara H, Bouchra O, Angéla FK, Samira EF, Nehemie N, Samir A. Acute rheumatic fever in children: Experience at the hospital Hassan II of Fez, Morocco. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2020. [DOI: 10.1016/j.cegh.2020.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
27
|
Washington A, Varki N, Valderrama JA, Nizet V, Bui JD. Evaluation of IL-17D in Host Immunity to Group A Streptococcus Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:3122-3129. [PMID: 33077643 DOI: 10.4049/jimmunol.1901482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
IL-17D is a cytokine that belongs to the IL-17 family and is conserved in vertebrates and invertebrates. In contrast to IL-17A and IL-17F, which are expressed in Th17 cells, IL-17D is expressed broadly in nonimmune cells. IL-17D can promote immune responses to cancer and viruses in part by inducing chemokines and recruiting innate immune cells such as NK cells. Although bacterial infection can induce IL-17D in fish and invertebrates, the role of mammalian IL-17D in antibacterial immunity has not been established. To determine whether IL-17D has a role in mediating host defense against bacterial infections, we studied i.p. infection by group A Streptococcus (GAS) in wild-type (WT) and Il17d -/- mice. Compared with WT animals, mice deficient in IL-17D experienced decreased survival, had greater weight loss, and showed increased bacterial burden in the kidney and peritoneal cavity following GAS challenge. In WT animals, IL-17D transcript was induced by GAS infection and correlated to increased levels of chemokine CCL2 and greater neutrophil recruitment. Of note, GAS-mediated IL-17D induction in nonimmune cells required live bacteria, suggesting that processes beyond recognition of pathogen-associated molecular patterns were required for IL-17D induction. Based on our results, we propose a model in which nonimmune cells can discriminate between nonviable and viable GAS cells, responding only to the latter by inducing IL-17D.
Collapse
Affiliation(s)
- Allen Washington
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Nissi Varki
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093
| | - J Andrés Valderrama
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92161; and
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92161; and.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Jack D Bui
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093;
| |
Collapse
|
28
|
Increasing incidence of group B streptococcus neonatal infections in the Netherlands is associated with clonal expansion of CC17 and CC23. Sci Rep 2020; 10:9539. [PMID: 32533007 PMCID: PMC7293262 DOI: 10.1038/s41598-020-66214-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/15/2020] [Indexed: 01/19/2023] Open
Abstract
Group B streptococcus (GBS) is the leading cause of neonatal invasive disease worldwide. In the Netherlands incidence of the disease increased despite implementation of preventive guidelines. We describe a genomic analysis of 1345 GBS isolates from neonatal (age 0–89 days) invasive infections in the Netherlands reported between 1987 and 2016. Most isolates clustered into one of five major lineages: CC17 (39%), CC19 (25%), CC23 (18%), CC10 (9%) and CC1 (7%). There was a significant rise in the number of infections due to isolates from CC17 and CC23. Phylogenetic clustering analysis revealed that this was caused by expansion of specific sub-lineages, designated CC17-A1, CC17-A2 and CC23-A1. Dating of phylogenetic trees estimated that these clones diverged in the 1960s/1970s, representing historical rather than recently emerged clones. For CC17-A1 the expansion correlated with acquisition of a new phage, carrying gene encoding a putative cell-surface protein. Representatives of CC17-A1, CC17-A2 and CC23-A1 clones were identified in datasets from other countries demonstrating their global distribution.
Collapse
|
29
|
Björck V, Påhlman LI, Bodelsson M, Petersson AC, Kander T. Morbidity and mortality in critically ill patients with invasive group A streptococcus infection: an observational study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:302. [PMID: 32505194 PMCID: PMC7275847 DOI: 10.1186/s13054-020-03008-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/20/2020] [Indexed: 12/23/2022]
Abstract
Background Group A streptococci (GAS) are known to cause serious invasive infections, but little is known about outcomes when patients with these infections are admitted to intensive care. We wanted to describe critically ill patients with severe sepsis or septic shock due to invasive GAS (iGAS) and compare them with other patients with severe sepsis or septic shock. Methods Adult patients admitted to a general intensive care unit (ICU) in Sweden (2007–2019) were screened for severe sepsis or septic shock according to Sepsis 2 definition. Individuals with iGAS infection were identified. The outcome variables were mortality, days alive and free of vasopressors and invasive mechanical ventilation, maximum acute kidney injury score for creatinine, use of continuous renal replacement therapy and maximum Sequential Organ Failure Assessment score during the ICU stay. Age, Simplified Acute Physiology Score (SAPS 3) and iGAS were used as independent, explanatory variables in regression analysis. Cox regression was used for survival analyses. Results iGAS was identified in 53 of 1021 (5.2%) patients. Patients with iGAS presented a lower median SAPS 3 score (62 [56–72]) vs 71 [61–81]), p < 0.001), had a higher frequency of cardiovascular cause of admission to the ICU (38 [72%] vs 145 [15%], p < 0.001) and had a higher median creatinine score (173 [100–311] vs 133 [86–208] μmol/L, p < 0.019). Of the GAS isolates, 50% were serotyped emm1/T1 and this group showed signs of more pronounced circulatory and renal failure than patients with non-emm1/T1 (p = 0.036 and p = 0.007, respectively). After correction for severity of illness (SAPS 3) and age, iGAS infection was associated with lower mortality risk (95% confidence interval (CI) of hazard ratio (HR) 0.204–0.746, p < 0.001). Morbidity analyses demonstrated that iGAS patients were more likely to develop renal failure. Conclusion Critically ill patients with iGAS infection had a lower mortality risk but a higher degree of renal failure compared to similarly ill sepsis patients. emm1/T1 was found to be the most dominant serotype, and patients with emm1/T1 demonstrated more circulatory and renal failure than patients with other serotypes of iGAS.
Collapse
Affiliation(s)
- Viveka Björck
- Skåne University Hospital, Department of Clinical Sciences Lund, Anaesthesiology and Intensive Care, Lund University, SE-221 85, Lund, Sweden.
| | - Lisa I Påhlman
- Skåne University Hospital, Department of Clinical Sciences Lund, Infection Medicine, Lund University, SE-221 85, Lund, Sweden
| | - Mikael Bodelsson
- Skåne University Hospital, Department of Clinical Sciences Lund, Anaesthesiology and Intensive Care, Lund University, SE-221 85, Lund, Sweden
| | | | - Thomas Kander
- Skåne University Hospital, Department of Clinical Sciences Lund, Anaesthesiology and Intensive Care, Lund University, SE-221 85, Lund, Sweden
| |
Collapse
|
30
|
Dissecting Streptococcus pyogenes interaction with human. Arch Microbiol 2020; 202:2023-2032. [PMID: 32504132 DOI: 10.1007/s00203-020-01932-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/26/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022]
Abstract
Streptococcus pyogenes is a species of Gram-positive bacteria. It is also known as Group A Streptococcus (GAS) that causes pathogenesis to humans only. The GAS infection has several manifestations including invasive illness. Current research has linked the molecular modes of GAS virulence with substantial sequencing determinations for the isolation of genomes. These advances help to comprehend the molecular evolution resulting in the pandemic strains. Thus, it is indispensable to reconsider the philosophy that involves GAS pathogenesis. The recent investigations involve studying GAS in the nasopharynx and its capability to cause infection or asymptomatically reside in the host. These advances have been discussed in this article with an emphasis on the natural history of GAS and the evolutionary change in the pandemic strains. In addition, this review describes the unique functions for major pathogenicity determinants to comprehend their physiological effects.
Collapse
|
31
|
Matsue M, Ogura K, Sugiyama H, Miyoshi-Akiyama T, Takemori-Sakai Y, Iwata Y, Wada T, Okamoto S. Pathogenicity Characterization of Prevalent-Type Streptococcus dysgalactiae subsp. equisimilis Strains. Front Microbiol 2020; 11:97. [PMID: 32117127 PMCID: PMC7010647 DOI: 10.3389/fmicb.2020.00097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/17/2020] [Indexed: 12/26/2022] Open
Abstract
Streptococcus dysgalactiae subsp. equisimilis (SDSE) is an emerging human pathogen that causes severe invasive streptococcal diseases. Recent reports have shown that SDSE exhibits high pathogenicity with different mechanisms from that of Streptococcus pyogenes, although the two streptococci possess some common virulence factors such as streptolysin, streptokinase, and cell-binding proteins. To date, only a few studies have examined the variety of mechanisms expressing the pathogenicity of SDSE. Among nine SDSE clinical isolates sequenced in this study, we present in vitro and in vivo analyses of KNZ01 and KNZ03, whose emm and multilocus species types (MLSTs) are prevalent in Japan and other countries. For the comparison of pathogenicity, we also utilized the ATCC 12394 strain. The whole-genome analysis showed that KNZ03 and ATCC 12394 are categorized into an identical clonal complex by MLST and are phylogenetically close. However, the three strains exhibited different characteristics for pathogenicity in vitro; ATCC 12394 showed significant cytotoxicity to human keratinocytes and release of streptolysin O (SLO) compared to KNZ01 and KNZ03; KNZ03 exhibited significantly high hemolytic activity, but did not secrete SLO. KNZ01 and KNZ03 adhered to human keratinocytes at a higher rate than ATCC 12394; KNZ03 showed a higher rate of survival after a brief (30 min) incubation with human neutrophils compared to the other two strains; also, KNZ01 grew more rapidly in the presence of human serum. In vivo subcutaneous infection commonly resulted in ulcer formation in the three strains 7 days after infection. KNZ01-infected mice showed significant body weight loss 2 days after infection. Besides, on post-infection day 2, only KNZ01 remained in the cutaneous tissues of mice. Scanning electron microscopy analysis revealed that KNZ01 formed an extracellular structure (biofilm), which was probably composed of cell wall-anchoring proteins, in the presence of glucose and human serum. The extracellular structure of ATCC 12394 was also changed dramatically in response to culture conditions, whereas that of KNZ03 did not. Our study proposed that each SDSE strain possesses different virulence factors characteristics for mediating pathogenicity in humans.
Collapse
Affiliation(s)
- Miki Matsue
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kohei Ogura
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Hironori Sugiyama
- Division of Instrumental Analysis, Engineering and Technology Department, Kanazawa University, Kanazawa, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Shinjuku, Japan
| | - Yukiko Takemori-Sakai
- Division of Clinical Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Division of Infection Control, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Division of Infection Control, Kanazawa University, Kanazawa, Japan.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Division of Clinical Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Shigefumi Okamoto
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
32
|
MUSSER JAMESM. MOLECULAR MECHANISMS CONTRIBUTING TO FUZZY EPIDEMICS CAUSED BY GROUP A STREPTOCOCCUS, A FLESH-EATING HUMAN BACTERIAL PATHOGEN. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2020; 131:356-368. [PMID: 32675873 PMCID: PMC7358509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Epidemics caused by microbial pathogens are inherently interesting because they can kill large numbers of our brethren, cause social upheaval, and alter history. Microbial epidemics will likely continue to occur at unpredictable times and result in poorly predictable consequences. Over a 30-year period, we have used the human bacterial pathogen group A streptococcus (also known as Streptococcus pyogenes) as a model organism to gain understanding of the molecular mechanisms contributing to epidemics caused by this pathogen and attendant virulence mechanisms. These epidemics have affected tens of millions of individuals worldwide and were largely unrecognized until revealed by full-genome sequence data from many thousands of isolates from intercontinental sources. Molecular genetic strategies, coupled with extensive use of relevant animal infection models, have delineated precise evolutionary genetic changes that contribute to pathogen clone emergence and successful dissemination among humans. Here, we summarize a few key findings from these studies.
Collapse
Affiliation(s)
- JAMES M. MUSSER
- Correspondence and reprint requests: James M. Musser, MD, PhD, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030713-441-5890713-441-3886
| |
Collapse
|
33
|
Streptococcus pyogenes Transcriptome Changes in the Inflammatory Environment of Necrotizing Fasciitis. Appl Environ Microbiol 2019; 85:AEM.01428-19. [PMID: 31471300 PMCID: PMC6803311 DOI: 10.1128/aem.01428-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/23/2019] [Indexed: 12/31/2022] Open
Abstract
Necrotizing fasciitis, a life-threatening subcutaneous soft-tissue infection, is principally caused by S. pyogenes. The inflammatory environment at the site of infection causes global gene expression changes for survival of the bacterium and pathogenesis. However, no known study regarding transcriptomic profiling of S. pyogenes in cases of necrotizing fasciitis has been presented. We identified 483 bacterial genes whose expression was consistently altered during infection. Our results showed that S. pyogenes infection induces drastic upregulation of the expression of virulence-associated genes and shifts metabolic pathway usage. In particular, high-level expression of toxins, such as cytolysins, proteases, and nucleases, was observed at infection sites. In addition, genes identified as consistently enriched included those related to metabolism of arginine and histidine as well as carbohydrate uptake and utilization. Conversely, genes associated with the oxidative stress response and cell division were consistently downregulated during infection. The present findings provide useful information for establishing novel treatment strategies. Streptococcus pyogenes is a major cause of necrotizing fasciitis, a life-threatening subcutaneous soft-tissue infection. At the host infection site, the local environment and interactions between the host and bacteria have effects on bacterial gene expression profiles, while the gene expression pattern of S. pyogenes related to this disease remains unknown. In this study, we used a mouse model of necrotizing fasciitis and performed RNA-sequencing (RNA-seq) analysis of S. pyogenes M1T1 strain 5448 by isolating total RNA from infected hind limbs obtained at 24, 48, and 96 h postinfection. RNA-seq analysis results identified 483 bacterial genes whose expression was consistently altered in the infected hindlimbs compared to their expression under in vitro conditions. Genes showing consistent enrichment during infection included 306 encoding molecules involved in virulence, carbohydrate utilization, amino acid metabolism, trace-metal transport, and the vacuolar ATPase transport system. Surprisingly, drastic upregulation of 3 genes, encoding streptolysin S precursor (sagA), cysteine protease (speB), and secreted DNase (spd), was noted in the present mouse model (log2 fold change, >6.0, >9.4, and >7.1, respectively). Conversely, the number of consistently downregulated genes was 177, including those associated with the oxidative stress response and cell division. These results suggest that in necrotizing fasciitis, S. pyogenes shows an altered metabolism, decreased cell proliferation, and upregulation of expression of major toxins. Our findings are considered to provide critical information for developing novel treatment strategies and vaccines for necrotizing fasciitis. IMPORTANCE Necrotizing fasciitis, a life-threatening subcutaneous soft-tissue infection, is principally caused by S. pyogenes. The inflammatory environment at the site of infection causes global gene expression changes for survival of the bacterium and pathogenesis. However, no known study regarding transcriptomic profiling of S. pyogenes in cases of necrotizing fasciitis has been presented. We identified 483 bacterial genes whose expression was consistently altered during infection. Our results showed that S. pyogenes infection induces drastic upregulation of the expression of virulence-associated genes and shifts metabolic pathway usage. In particular, high-level expression of toxins, such as cytolysins, proteases, and nucleases, was observed at infection sites. In addition, genes identified as consistently enriched included those related to metabolism of arginine and histidine as well as carbohydrate uptake and utilization. Conversely, genes associated with the oxidative stress response and cell division were consistently downregulated during infection. The present findings provide useful information for establishing novel treatment strategies.
Collapse
|
34
|
O. Luiz FBD, Alves KB, Barros RR. Prevalence and long-term persistence of beta-haemolytic streptococci throat carriage among children and young adults. J Med Microbiol 2019; 68:1526-1533. [DOI: 10.1099/jmm.0.001054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Fernanda Baptista de O. Luiz
- Departamento Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Rua Professor Ernani de Melo 101, Niterói, RJ, 24210-130, Brazil
| | - Karen B. Alves
- Departamento Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Rua Professor Ernani de Melo 101, Niterói, RJ, 24210-130, Brazil
| | - Rosana R. Barros
- Departamento Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Rua Professor Ernani de Melo 101, Niterói, RJ, 24210-130, Brazil
| |
Collapse
|
35
|
Brouwer S, Lacey JA, You Y, Davies MR, Walker MJ. Scarlet fever changes its spots. THE LANCET. INFECTIOUS DISEASES 2019; 19:1154-1155. [PMID: 31519542 DOI: 10.1016/s1473-3099(19)30494-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/05/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Stephan Brouwer
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jake A Lacey
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Yuanhai You
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mark R Davies
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
36
|
Li Y, Liu L, Huang Z, Yang L, Ye Y, Li R. A case of streptococcus necrotizing fasciitis secondary to acute tonsillitis and review of literatures. HONG KONG J EMERG ME 2019. [DOI: 10.1177/1024907918767190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Necrotizing fasciitis, officially named by Wilson in 1952, was a rare potentially life-threatening necrotizing soft tissue infections disease. The main lesions showed subcutaneous fat and fascia layer in progressive necrosis, generally not involving muscles. It was characterized by acute onset, rapid progress, and dangerous condition, often accompanied by systemic toxin shock, which was a relatively rare clinical acute critical disease with high mortality rate. Necrotizing fasciitis is a rare but clinical critical disease. The overall incidence of necrotizing fasciitis is approximately 0.04 cases per 1000 persons in the United States, the 30-day mortality is 27%, and necrotizing fasciitis–related mortality in Asian region is about 28%. Streptococcus pyogenes (group A streptococcus) are human-specific pathogens that can cause upper respiratory tract infection such as tonsillitis, associated with post-infection diseases such as rheumatic fever, and also can induce severe invasive diseases such as necrotizing fasciitis and streptococcus toxin shock syndrome. This article reports a case of streptococcus necrotizing fasciitis secondary to suppurative tonsillitis, which was seldom reported before and our successful management with delayed debridement. This study was anonymous and was approved by the local Research Ethics Committee. Informed consent was obtained from the patient.
Collapse
Affiliation(s)
- Yongsheng Li
- Department of Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Lu Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Zhonghui Huang
- Department of Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Le Yang
- Department of Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yan Ye
- Department of Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Renjie Li
- Department of Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
37
|
Coppens J, Xavier BB, Loens K, Lammens C, Ieven M, Matheeussen V, Goossens H, Malhotra-Kumar S. Remarkable Genome Stability among emm1 Group A Streptococcus in Belgium over 19 Years. Genome Biol Evol 2019; 11:1432-1439. [PMID: 31065672 PMCID: PMC6521816 DOI: 10.1093/gbe/evz093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2019] [Indexed: 12/30/2022] Open
Abstract
During the last two decades, there has been a public health concern of severe invasive infections caused by Group A Streptococcus (GAS) of the emm1 genotype. This study investigated the dynamics of emm1 GAS during 1994–2013 in Belgium. emm1 GAS isolated from blood, tissue, and wounds of patients with invasive infections (n = 23, S1–S23), and from patients with uncomplicated pharyngitis (n = 15, NS1–NS15) were subjected to whole-genome mapping (WGM; kpn) (Opgen). Whole-genome sequencing was performed on 25 strains (WGS; S1–S23 and NS6–NS7) (Illumina Inc.). Belgian GAS belonged to the M1T1 clone typified by the 36-kb chromosomal region encoding extracellular toxins, NAD+-glycohydrolase and streptolysin O. Strains from 1994–1999 clustered together with published strains (MGAS5005 and M1476). From 2001 onward, invasive GAS showed higher genomic divergence in the accessory genome and harbored on average 7% prophage content. Low evolutionary rate (2.49E-008; P > 0.05) was observed in this study, indicating a highly stable genome. The studied invasive and pharyngitis isolates were no genetically distinct populations based on the WGM and core genome phylogeny analyses. Two copies of the speJ superantigen were present in the 1999 and 2010 study strains (n = 3), one being chromosomal and one being truncated and associated with phage remnants. This study showed that emm1 GAS in Belgium, compared with Canada and UK M1 strains, were highly conserved by harboring a remarkable genome stability over a 19-year period with variations observed in the accessory genome.
Collapse
Affiliation(s)
- Jasmine Coppens
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| | - Basil Britto Xavier
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| | - Katherine Loens
- Belgian Reference Centre for Group A Streptococcus, Antwerp University Hospital, Antwerp, Belgium
| | - Christine Lammens
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| | - Margareta Ieven
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium.,Belgian Reference Centre for Group A Streptococcus, Antwerp University Hospital, Antwerp, Belgium
| | - Veerle Matheeussen
- Belgian Reference Centre for Group A Streptococcus, Antwerp University Hospital, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium.,Belgian Reference Centre for Group A Streptococcus, Antwerp University Hospital, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| |
Collapse
|
38
|
McNitt DH, Van De Water L, Marasco D, Berisio R, Lukomski S. Streptococcal Collagen-like Protein 1 Binds Wound Fibronectin: Implications in Pathogen Targeting. Curr Med Chem 2019; 26:1933-1945. [PMID: 30182848 DOI: 10.2174/0929867325666180831165704] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/18/2018] [Accepted: 06/28/2018] [Indexed: 02/01/2023]
Abstract
Group A Streptococcus (GAS) infections are responsible for significant morbidity and mortality worldwide. The outlook for an effective global vaccine is reduced because of significant antigenic variation among GAS strains worldwide. Other challenges in GAS therapy include the lack of common access to antibiotics in developing countries, as well as allergy to and treatment failures with penicillin and increasing erythromycin resistance in the industrialized world. At the portal of entry, GAS binds to newly deposited extracellular matrix, which is rich in cellular fibronectin isoforms with extra domain A (EDA, also termed EIIIA) via the surface adhesin, the streptococcal collagen-like protein 1 (Scl1). Recombinant Scl1 constructs, derived from diverse GAS strains, bind the EDA loop segment situated between the C and C' β-strands. Despite the sequence diversity in Scl1 proteins, multiple sequence alignments and secondary structure predictions of Scl1 variants, as well as crystallography and homology modeling studies, point to a conserved mechanism of Scl1-EDA binding. We propose that targeting this interaction may prevent the progression of infection. A synthetic cyclic peptide, derived from the EDA C-C' loop, binds to recombinant Scl1 with a micromolar dissociation constant. This review highlights the current concept of EDA binding to Scl1 and provides incentives to exploit this binding to treat GAS infections and wound colonization.
Collapse
Affiliation(s)
- Dudley H McNitt
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, 2095 Health Sciences North, Morgantown, WV 26506, United States
| | - Livingston Van De Water
- Departments of Surgery and Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, United States
| | - Daniela Marasco
- Department of Pharmacy, University of Naples Frederico II, Naples, Italy
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council, via Mezzocannone, 16, 80134, Naples, Italy
| | - Slawomir Lukomski
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, 2095 Health Sciences North, Morgantown, WV 26506, United States
| |
Collapse
|
39
|
De Oliveira DMP, Everest-Dass A, Hartley-Tassell L, Day CJ, Indraratna A, Brouwer S, Cleary A, Kautto L, Gorman J, Packer NH, Jennings MP, Walker MJ, Sanderson-Smith ML. Human glycan expression patterns influence Group A streptococcal colonization of epithelial cells. FASEB J 2019; 33:10808-10818. [PMID: 31262188 DOI: 10.1096/fj.201900559r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Colonization of the oropharynx is the initial step in Group A Streptococcus (GAS) pharyngeal infection. We have previously reported that the highly virulent M1T1 GAS clone attaches to oral epithelial cells via M1 protein interaction with blood group antigen carbohydrate structures. Here, we have identified that colonization of human oral epithelial cells by GAS serotypes M3 and M12 is mediated by human blood group antigens [ABO(H)] and Lewis (Le) antigen expression. Removal of linkage-specific fucose, galactose, N-acetylgalactosamine, and sialic acid modulated GAS colonization, dependent on host ABO(H) blood group and Le expression profile. Furthermore, N-linked glycans from human salivary glycoproteins, when released and purified, were potent inhibitors of M1, M3, and M12 GAS colonization ex vivo. These data highlight the important role played by human protein glycosylation patterns in GAS attachment to oral epithelial cell surfaces.-De Oliveira, D. M. P., Everest-Dass, A., Hartley-Tassell, L., Day, C. J., Indraratna, A., Brouwer, S., Cleary, A., Kautto, L., Gorman, J., Packer, N. H., Jennings, M. P., Walker, M. J., Sanderson-Smith, M. L. Human glycan expression patterns influence Group A streptococcal colonization of epithelial cells.
Collapse
Affiliation(s)
- David M P De Oliveira
- School of Chemistry and Molecular Bioscience, Molecular Horizons and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, Australia.,Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Arun Everest-Dass
- Department of Molecular Sciences, Macquarie University, Sydney, New South Wales, Australia.,Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | | | - Christopher J Day
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Anuk Indraratna
- School of Chemistry and Molecular Bioscience, Molecular Horizons and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Stephan Brouwer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Ailish Cleary
- School of Chemistry and Molecular Bioscience, Molecular Horizons and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Liisa Kautto
- Department of Molecular Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Jody Gorman
- School of Chemistry and Molecular Bioscience, Molecular Horizons and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Nicolle H Packer
- Department of Molecular Sciences, Macquarie University, Sydney, New South Wales, Australia.,Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Michael P Jennings
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Mark J Walker
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Martina L Sanderson-Smith
- School of Chemistry and Molecular Bioscience, Molecular Horizons and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, Australia
| |
Collapse
|
40
|
Freydlin IS, Starikova EA, Lebedeva AM. Overcoming the protective functions of macrophages by Streptococcus pyogenes virulence factors. BULLETIN OF SIBERIAN MEDICINE 2019. [DOI: 10.20538/1682-0363-2019-1-109-118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The review is devoted to the analysis of molecular mechanisms of action ofS. pyogenesvirulence factors aimed at overcoming the protective functions of macrophages. The review describes in detail the main protective functions of macrophages and the mechanisms of their implementation in the course of streptococcal infection. The virulence factors ofS. pyogenes,which prevent the recruitment of macrophages to the site of infection, are examined. Particular attention is paid to the analysis of molecular effects that suppress the pathogen by the process of phagocytosis, intracellular bactericidal activity and the production of cytokines by macrophages. The analysis of molecular genetic mechanisms of regulation of the expression ofS. pyogenesvirulence factors that provide adaptation of the pathogen to changing conditions in the site of inflammation is carried out.
Collapse
Affiliation(s)
- I. S. Freydlin
- Institute of Experimental Medicine; Pavlov First Saint Petersburg State Medical University
| | | | | |
Collapse
|
41
|
Rivera-Hernandez T, Carnathan DG, Jones S, Cork AJ, Davies MR, Moyle PM, Toth I, Batzloff MR, McCarthy J, Nizet V, Goldblatt D, Silvestri G, Walker MJ. An Experimental Group A Streptococcus Vaccine That Reduces Pharyngitis and Tonsillitis in a Nonhuman Primate Model. mBio 2019; 10:e00693-19. [PMID: 31040243 PMCID: PMC6495378 DOI: 10.1128/mbio.00693-19] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022] Open
Abstract
Group A Streptococcus (GAS) infections account for an estimated 500,000 deaths every year. This bacterial pathogen is responsible for a variety of mild and life-threatening infections and the triggering of chronic autoimmune sequelae. Pharyngitis caused by group A Streptococcus (GAS), but not asymptomatic GAS carriage, is a prerequisite for acute rheumatic fever (ARF). Repeated bouts of ARF may trigger rheumatic heart disease (RHD), a major cause of heart failure and stroke accounting for 275,000 deaths annually. A vaccine that prevents pharyngitis would markedly reduce morbidity and mortality from ARF and RHD. Nonhuman primates (NHPs) have been utilized to model GAS diseases, and experimentally infected rhesus macaques develop pharyngitis. Here we use an NHP model of GAS pharyngitis to evaluate the efficacy of an experimental vaccine, Combo5 (arginine deiminase [ADI], C5a peptidase [SCPA], streptolysin O [SLO], interleukin-8 [IL-8] protease [SpyCEP], and trigger factor [TF]), specifically designed to exclude GAS components potentially linked to autoimmune complications. Antibody responses against all Combo5 antigens were detected in NHP serum, and immunized NHPs showed a reduction in pharyngitis and tonsillitis compared to controls. Our work establishes the NHP model as a gold standard for the assessment of GAS vaccines.IMPORTANCE GAS-related diseases disproportionally affect disadvantaged populations (e.g., indigenous populations), and development of a vaccine has been neglected. A recent strong advocacy campaign driven by the World Health Organization and the International Vaccine Institute has highlighted the urgent need for a GAS vaccine. One significant obstacle in GAS vaccine development is the lack of a widely used animal model to assess vaccine efficacy. Researchers in the field use a wide range of murine models of infection and in vitro assays, sometimes yielding conflicting results. Here we present the nonhuman primate pharyngeal infection model as a tool to assess vaccine-induced protection against colonization and clinical symptoms of pharyngitis and tonsillitis. We have tested the efficacy of an experimental vaccine candidate with promising results. We believe that the utilization of this valuable tool by the GAS vaccine research community could significantly accelerate the realization of a safe and effective GAS vaccine for humans.
Collapse
Affiliation(s)
- Tania Rivera-Hernandez
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Diane G Carnathan
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Scott Jones
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Amanda J Cork
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mark R Davies
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
- Peter Doherty Institute, University of Melbourne, Parkville, VIC, Australia
| | - Peter M Moyle
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
- School of Pharmacy, The University of Queensland, St Lucia, QLD, Australia
| | - Istvan Toth
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Michael R Batzloff
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - James McCarthy
- Australian Infectious Diseases Research Centre, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla, California, USA
| | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Guido Silvestri
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Mark J Walker
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
42
|
Dan JM, Havenar-Daughton C, Kendric K, Al-Kolla R, Kaushik K, Rosales SL, Anderson EL, LaRock CN, Vijayanand P, Seumois G, Layfield D, Cutress RI, Ottensmeier CH, Lindestam Arlehamn CS, Sette A, Nizet V, Bothwell M, Brigger M, Crotty S. Recurrent group A Streptococcus tonsillitis is an immunosusceptibility disease involving antibody deficiency and aberrant T FH cells. Sci Transl Med 2019; 11:eaau3776. [PMID: 30728285 PMCID: PMC6561727 DOI: 10.1126/scitranslmed.aau3776] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/29/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022]
Abstract
"Strep throat" is highly prevalent among children, yet it is unknown why only some children develop recurrent tonsillitis (RT), a common indication for tonsillectomy. To gain insights into this classic childhood disease, we performed phenotypic, genotypic, and functional studies on pediatric group A Streptococcus (GAS) RT and non-RT tonsils from two independent cohorts. GAS RT tonsils had smaller germinal centers, with an underrepresentation of GAS-specific CD4+ germinal center T follicular helper (GC-TFH) cells. RT children exhibited reduced antibody responses to an important GAS virulence factor, streptococcal pyrogenic exotoxin A (SpeA). Risk and protective human leukocyte antigen (HLA) class II alleles for RT were identified. Lastly, SpeA induced granzyme B production in GC-TFH cells from RT tonsils with the capacity to kill B cells and the potential to hobble the germinal center response. These observations suggest that RT is a multifactorial disease and that contributors to RT susceptibility include HLA class II differences, aberrant SpeA-activated GC-TFH cells, and lower SpeA antibody titers.
Collapse
Affiliation(s)
- Jennifer M Dan
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Colin Havenar-Daughton
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), La Jolla, CA 92037, USA
| | - Kayla Kendric
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Rita Al-Kolla
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Kirti Kaushik
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Sandy L Rosales
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Ericka L Anderson
- Department of Pediatrics, School of Medicine, UCSD, La Jolla, CA 92037, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA 92037, USA
- Human Longevity Inc., San Diego, CA 92121, USA
| | - Christopher N LaRock
- Department of Pediatrics, School of Medicine, UCSD, La Jolla, CA 92037, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA 92037, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Pandurangan Vijayanand
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Grégory Seumois
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - David Layfield
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, UK
| | - Ramsey I Cutress
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, UK
| | | | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Victor Nizet
- Department of Pediatrics, School of Medicine, UCSD, La Jolla, CA 92037, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA 92037, USA
| | - Marcella Bothwell
- Division of Pediatric Otolaryngology, Rady Children's Hospital, San Diego, CA 92123, USA
- Department of Surgery, UCSD, La Jolla, CA 92037, USA
- Department of Otolaryngology, Head and Neck Surgery, Naval Medical Center San Diego, San Diego, CA 92134, USA
| | - Matthew Brigger
- Division of Pediatric Otolaryngology, Rady Children's Hospital, San Diego, CA 92123, USA
- Department of Surgery, UCSD, La Jolla, CA 92037, USA
- Department of Otolaryngology, Head and Neck Surgery, Naval Medical Center San Diego, San Diego, CA 92134, USA
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
- Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), La Jolla, CA 92037, USA
| |
Collapse
|
43
|
Group A Streptococcal DNase Sda1 Impairs Plasmacytoid Dendritic Cells' Type 1 Interferon Response. J Invest Dermatol 2018; 139:1284-1293. [PMID: 30543898 DOI: 10.1016/j.jid.2018.11.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 01/19/2023]
Abstract
Group A Streptococcus causes severe invasive infections, including necrotizing fasciitis. The expression of an array of virulence factors targeting specific host immune functions impedes successful bacterial clearance. The virulence factor streptococcal DNase Sda1 was previously shown to interfere with the entrapment of bacteria through neutrophil extracellular traps and TLR9 signaling. In this study, we showed that plasmacytoid dendritic cells are recruited to the infected tissue during group A streptococcal necrotizing fasciitis. We found that the streptococcal DNase Sda1 impairs plasmacytoid dendritic cell recruitment by reducing IFN-1 levels at the site of infection. We found that streptococcal DNase Sda1 interferes with stabilization of the DNA by the host molecule HMGB1 protein, which may account for decreased IFN-1 levels at the site of infection.
Collapse
|
44
|
Pato C, Melo-Cristino J, Ramirez M, Friães A. Streptococcus pyogenes Causing Skin and Soft Tissue Infections Are Enriched in the Recently Emerged emm89 Clade 3 and Are Not Associated With Abrogation of CovRS. Front Microbiol 2018; 9:2372. [PMID: 30356787 PMCID: PMC6189468 DOI: 10.3389/fmicb.2018.02372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/18/2018] [Indexed: 11/29/2022] Open
Abstract
Although skin and soft tissue infections (SSTI) are the most common focal infections associated with invasive disease caused by Streptococcus pyogenes (Lancefield Group A streptococci - GAS), there is scarce information on the characteristics of isolates recovered from SSTI in temperate-climate regions. In this study, 320 GAS isolated from SSTI in Portugal were characterized by multiple typing methods and tested for antimicrobial susceptibility and SpeB activity. The covRS and ropB genes of isolates with no detectable SpeB activity were sequenced. The antimicrobial susceptibility profile was similar to that of previously characterized isolates from invasive infections (iGAS), presenting a decreasing trend in macrolide resistance. However, the clonal composition of SSTI between 2005 and 2009 was significantly different from that of contemporary iGAS. Overall, iGAS were associated with emm1 and emm3, while SSTI were associated with emm89, the dominant emm type among SSTI (19%). Within emm89, SSTI were only significantly associated with isolates lacking the hasABC locus, suggesting that the recently emerged emm89 clade 3 may have an increased potential to cause SSTI. Reflecting these associations between emm type and disease presentation, there were also differences in the distribution of emm clusters, sequence types, and superantigen gene profiles between SSTI and iGAS. According to the predicted ability of each emm cluster to interact with host proteins, iGAS were associated with the ability to bind fibrinogen and albumin, whereas SSTI isolates were associated with the ability to bind C4BP, IgA, and IgG. SpeB activity was absent in 79 isolates (25%), in line with the proportion previously observed among iGAS. Null covS and ropB alleles (predicted to eliminate protein function) were detected in 10 (3%) and 12 (4%) isolates, corresponding to an underrepresentation of mutations impairing CovRS function in SSTI relative to iGAS. Overall, these results indicate that the isolates responsible for SSTI are genetically distinct from those recovered from normally sterile sites, supporting a role for mutations impairing CovRS activity specifically in invasive infection and suggesting that this role relies on a differential regulation of other virulence factors besides SpeB.
Collapse
Affiliation(s)
- Catarina Pato
- Author Affiliations: Centro Hospitalar do Barlavento Algarvio; Centro Hospitalar de Entre Douro e Vouga; Centro Hospitalar de Leiria; Centro Hospitalar de Vila Nova de Gaia/Espinho; Centro Hospitalar do Alto Ave; Centro Hospitalar do Porto; Centro Hospitalar da Póvoa do Varzim/Vila do Conde; Hospital Central do Funchal; Centro Hospitalar de Lisboa Central; Centro Hospitalar Lisboa Norte; Centro Hospitalar Lisboa Ocidental; Centro Hospitalar do Baixo Vouga; Hospital de Vila Real; Hospitais da Universidade de Coimbra; Hospital de Cascais; Hospital de São João, Porto; Hospital de Braga; Hospital de Santa Luzia, Elvas; Hospital dos SAMS, Lisboa; Hospital Dr. Fernando da Fonseca, Amadora/Sintra; Hospital do Espírito Santo, Évora; Hospital Garcia de Orta, Almada; Hospital Pedro Hispano, Matosinhos; Unidade Local de Saúde do Baixo Alentejo, Beja.,Faculdade de Medicina, Instituto de Microbiologia, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - José Melo-Cristino
- Author Affiliations: Centro Hospitalar do Barlavento Algarvio; Centro Hospitalar de Entre Douro e Vouga; Centro Hospitalar de Leiria; Centro Hospitalar de Vila Nova de Gaia/Espinho; Centro Hospitalar do Alto Ave; Centro Hospitalar do Porto; Centro Hospitalar da Póvoa do Varzim/Vila do Conde; Hospital Central do Funchal; Centro Hospitalar de Lisboa Central; Centro Hospitalar Lisboa Norte; Centro Hospitalar Lisboa Ocidental; Centro Hospitalar do Baixo Vouga; Hospital de Vila Real; Hospitais da Universidade de Coimbra; Hospital de Cascais; Hospital de São João, Porto; Hospital de Braga; Hospital de Santa Luzia, Elvas; Hospital dos SAMS, Lisboa; Hospital Dr. Fernando da Fonseca, Amadora/Sintra; Hospital do Espírito Santo, Évora; Hospital Garcia de Orta, Almada; Hospital Pedro Hispano, Matosinhos; Unidade Local de Saúde do Baixo Alentejo, Beja.,Faculdade de Medicina, Instituto de Microbiologia, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Mario Ramirez
- Author Affiliations: Centro Hospitalar do Barlavento Algarvio; Centro Hospitalar de Entre Douro e Vouga; Centro Hospitalar de Leiria; Centro Hospitalar de Vila Nova de Gaia/Espinho; Centro Hospitalar do Alto Ave; Centro Hospitalar do Porto; Centro Hospitalar da Póvoa do Varzim/Vila do Conde; Hospital Central do Funchal; Centro Hospitalar de Lisboa Central; Centro Hospitalar Lisboa Norte; Centro Hospitalar Lisboa Ocidental; Centro Hospitalar do Baixo Vouga; Hospital de Vila Real; Hospitais da Universidade de Coimbra; Hospital de Cascais; Hospital de São João, Porto; Hospital de Braga; Hospital de Santa Luzia, Elvas; Hospital dos SAMS, Lisboa; Hospital Dr. Fernando da Fonseca, Amadora/Sintra; Hospital do Espírito Santo, Évora; Hospital Garcia de Orta, Almada; Hospital Pedro Hispano, Matosinhos; Unidade Local de Saúde do Baixo Alentejo, Beja.,Faculdade de Medicina, Instituto de Microbiologia, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Friães
- Author Affiliations: Centro Hospitalar do Barlavento Algarvio; Centro Hospitalar de Entre Douro e Vouga; Centro Hospitalar de Leiria; Centro Hospitalar de Vila Nova de Gaia/Espinho; Centro Hospitalar do Alto Ave; Centro Hospitalar do Porto; Centro Hospitalar da Póvoa do Varzim/Vila do Conde; Hospital Central do Funchal; Centro Hospitalar de Lisboa Central; Centro Hospitalar Lisboa Norte; Centro Hospitalar Lisboa Ocidental; Centro Hospitalar do Baixo Vouga; Hospital de Vila Real; Hospitais da Universidade de Coimbra; Hospital de Cascais; Hospital de São João, Porto; Hospital de Braga; Hospital de Santa Luzia, Elvas; Hospital dos SAMS, Lisboa; Hospital Dr. Fernando da Fonseca, Amadora/Sintra; Hospital do Espírito Santo, Évora; Hospital Garcia de Orta, Almada; Hospital Pedro Hispano, Matosinhos; Unidade Local de Saúde do Baixo Alentejo, Beja.,Faculdade de Medicina, Instituto de Microbiologia, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | | |
Collapse
|
45
|
Hertzog BB, Kaufman Y, Biswas D, Ravins M, Ambalavanan P, Wiener R, Angeli V, Chen SL, Hanski E. A Sub-population of Group A Streptococcus Elicits a Population-wide Production of Bacteriocins to Establish Dominance in the Host. Cell Host Microbe 2018; 23:312-323.e6. [PMID: 29544095 DOI: 10.1016/j.chom.2018.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/26/2017] [Accepted: 02/07/2018] [Indexed: 11/30/2022]
Abstract
Bacteria use quorum sensing (QS) to regulate gene expression. We identified a group A Streptococcus (GAS) strain possessing the QS system sil, which produces functional bacteriocins, through a sequential signaling pathway integrating host and bacterial signals. Host cells infected by GAS release asparagine (ASN), which is sensed by the bacteria to alter its gene expression and rate of proliferation. We show that upon ASN sensing, GAS upregulates expression of the QS autoinducer peptide SilCR. Initial SilCR expression activates the autoinduction cycle for further SilCR production. The autoinduction process propagates throughout the GAS population, resulting in bacteriocin production. Subcutaneous co-injection of mice with a bacteriocin-producing strain and the globally disseminated M1T1 GAS clone results in M1T1 killing within soft tissue. Thus, by sensing host signals, a fraction of a bacterial population can trigger an autoinduction mechanism mediated by QS, which acts on the entire bacterial community to outcompete other bacteria within the infection.
Collapse
Affiliation(s)
- Baruch B Hertzog
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 9112102, Israel
| | - Yael Kaufman
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 9112102, Israel
| | - Debabrata Biswas
- NUS-HUJ-CREATE Programme for Inflammation Research, Center for Research Excellence & Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| | - Miriam Ravins
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 9112102, Israel
| | - Poornima Ambalavanan
- NUS-HUJ-CREATE Programme for Inflammation Research, Center for Research Excellence & Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore
| | - Reuven Wiener
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 9112102, Israel
| | - Veronique Angeli
- Department of Microbiology and Immunology, National University of Singapore; LSI Immunology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Swaine L Chen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, and Infectious Diseases Group, Genome Institute of Singapore, Singapore 119074, Singapore
| | - Emanuel Hanski
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 9112102, Israel; NUS-HUJ-CREATE Programme for Inflammation Research, Center for Research Excellence & Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore, Singapore 138602, Singapore.
| |
Collapse
|
46
|
Ermert D, Weckel A, Magda M, Mörgelin M, Shaughnessy J, Rice PA, Björck L, Ram S, Blom AM. Human IgG Increases Virulence of Streptococcus pyogenes through Complement Evasion. THE JOURNAL OF IMMUNOLOGY 2018; 200:3495-3505. [PMID: 29626087 DOI: 10.4049/jimmunol.1800090] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022]
Abstract
Streptococcus pyogenes is an exclusively human pathogen that can provoke mild skin and throat infections but can also cause fatal septicemia. This gram-positive bacterium has developed several strategies to evade the human immune system, enabling S. pyogenes to survive in the host. These strategies include recruiting several human plasma proteins, such as the complement inhibitor, C4b-binding protein (C4BP), and human (hu)-IgG through its Fc region to the bacterial surface to evade immune recognition. We identified a novel virulence mechanism whereby IgG-enhanced binding of C4BP to five of 12 tested S. pyogenes strains expressed diverse M proteins that are important surface-expressed virulence factors. Importantly, all strains that bound C4BP in the absence of IgG bound more C4BP when IgG was present. Further studies with an M1 strain that additionally expressed protein H, also a member of the M protein family, revealed that binding of hu-IgG Fc to protein H increased the affinity of protein H for C4BP. Increased C4BP binding accentuated complement downregulation, resulting in diminished bacterial killing. Accordingly, mortality from S. pyogenes infection in hu-C4BP transgenic mice was increased when hu-IgG or its Fc portion alone was administered concomitantly. Electron microscopy analysis of human tissue samples with necrotizing fasciitis confirmed increased C4BP binding to S. pyogenes when IgG was present. Our findings provide evidence of a paradoxical function of hu-IgG bound through Fc to diverse S. pyogenes isolates that increases their virulence and may counteract the beneficial effects of IgG opsonization.
Collapse
Affiliation(s)
- David Ermert
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214 28 Malmo, Sweden; .,Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Antonin Weckel
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214 28 Malmo, Sweden
| | - Michal Magda
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214 28 Malmo, Sweden
| | - Matthias Mörgelin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Jutamas Shaughnessy
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Peter A Rice
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Lars Björck
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, 214 28 Malmo, Sweden
| |
Collapse
|
47
|
Endopeptidase PepO Regulates the SpeB Cysteine Protease and Is Essential for the Virulence of Invasive M1T1 Streptococcus pyogenes. J Bacteriol 2018; 200:JB.00654-17. [PMID: 29378883 DOI: 10.1128/jb.00654-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/18/2018] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus [GAS]) causes a wide range of human infections. The pathogenesis of GAS infections is dependent on the temporal expression of numerous secreted and surface-associated virulence factors that interact with host proteins. Streptococcal pyrogenic exotoxin B (SpeB) is one of the most extensively studied toxins produced by GAS, and the coordinate growth phase-dependent regulation of speB expression is linked to disease severity phenotypes. Here, we identified the endopeptidase PepO as a novel growth phase-dependent regulator of SpeB in the invasive GAS M1 serotype strain 5448. By using transcriptomics followed by quantitative reverse transcriptase PCR and Western blot analyses, we demonstrate through targeted mutagenesis that PepO influences growth phase-dependent induction of speB gene expression. Compared to wild-type and complemented mutant strains, we demonstrate that the 5448ΔpepO mutant strain is more susceptible to killing by human neutrophils and is attenuated in virulence in a murine model of invasive GAS infection. Our results expand the complex regulatory network that is operating in GAS to control SpeB production and suggest that PepO is a virulence requirement during GAS M1T1 strain 5448 infections.IMPORTANCE Despite the continuing susceptibility of S. pyogenes to penicillin, this bacterial pathogen remains a leading infectious cause of global morbidity and mortality. A particular subclone of the M1 serotype (M1T1) has persisted globally for decades as the most frequently isolated serotype from patients with invasive and noninvasive diseases in Western countries. One of the key GAS pathogenicity factors is the potent broad-spectrum cysteine protease SpeB. Although there has been extensive research interest on the regulatory mechanisms that control speB gene expression, its genetic regulation is not fully understood. Here, we identify the endopeptidase PepO as a new regulator of speB gene expression in the globally disseminated M1T1 clone and as being essential for virulence.
Collapse
|
48
|
Clark M, Kim J, Etesami N, Shimamoto J, Whalen RV, Martin G, Okumura CYM. Group A Streptococcus Prevents Mast Cell Degranulation to Promote Extracellular Trap Formation. Front Immunol 2018; 9:327. [PMID: 29535718 PMCID: PMC5835080 DOI: 10.3389/fimmu.2018.00327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/06/2018] [Indexed: 12/30/2022] Open
Abstract
The resurgence of Group A Streptococcus (GAS) infections in the past two decades has been a rising major public health concern. Due to a large number of GAS infections occurring in the skin, mast cells (MCs), innate immune cells known to localize to the dermis, could play an important role in controlling infection. MCs can exert their antimicrobial activities either early during infection, by degranulation and release of antimicrobial proteases and the cathelicidin-derived antimicrobial peptide LL-37, or by forming antibacterial MC extracellular traps (MCETs) in later stages of infection. We demonstrate that MCs do not directly degranulate in response to GAS, reducing their ability to control bacterial growth in early stages of infection. However, MC granule components are highly cytotoxic to GAS due to the pore-forming activity of LL-37, while MC granule proteases do not significantly affect GAS viability. We therefore confirmed the importance of MCETs by demonstrating their capacity to reduce GAS survival. The data therefore suggests that LL-37 from MC granules become embedded in MCETs, and are the primary effector molecule by which MCs control GAS infection. Our work underscores the importance of a non-traditional immune effector cell, utilizing a non-conventional mechanism, in the defense against an important human pathogen.
Collapse
Affiliation(s)
- Mary Clark
- Department of Biology, Occidental College, Los Angeles, CA, United States
| | - Jessica Kim
- Department of Biology, Occidental College, Los Angeles, CA, United States
| | - Neelou Etesami
- Department of Biology, Occidental College, Los Angeles, CA, United States
| | | | - Ryan V. Whalen
- Department of Biology, Occidental College, Los Angeles, CA, United States
| | - Gary Martin
- Department of Biology, Occidental College, Los Angeles, CA, United States
| | | |
Collapse
|
49
|
Hughes D, Andersson DI. Environmental and genetic modulation of the phenotypic expression of antibiotic resistance. FEMS Microbiol Rev 2018; 41:374-391. [PMID: 28333270 PMCID: PMC5435765 DOI: 10.1093/femsre/fux004] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/01/2017] [Indexed: 12/22/2022] Open
Abstract
Antibiotic resistance can be acquired by mutation or horizontal transfer of a resistance gene, and generally an acquired mechanism results in a predictable increase in phenotypic resistance. However, recent findings suggest that the environment and/or the genetic context can modify the phenotypic expression of specific resistance genes/mutations. An important implication from these findings is that a given genotype does not always result in the expected phenotype. This dissociation of genotype and phenotype has important consequences for clinical bacteriology and for our ability to predict resistance phenotypes from genetics and DNA sequences. A related problem concerns the degree to which the genes/mutations currently identified in vitro can fully explain the in vivo resistance phenotype, or whether there is a significant additional amount of presently unknown mutations/genes (genetic ‘dark matter’) that could contribute to resistance in clinical isolates. Finally, a very important question is whether/how we can identify the genetic features that contribute to making a successful pathogen, and predict why some resistant clones are very successful and spread globally? In this review, we describe different environmental and genetic factors that influence phenotypic expression of antibiotic resistance genes/mutations and how this information is needed to understand why particular resistant clones spread worldwide and to what extent we can use DNA sequences to predict evolutionary success.
Collapse
Affiliation(s)
- Diarmaid Hughes
- Corresponding author: Department of Medical Biochemistry and Microbiology, Biomedical Center (Box 582), Uppsala University, S-751 23 Uppsala, Sweden. Tel: +46 18 4714507; E-mail:
| | | |
Collapse
|
50
|
Abstract
Group A Streptococcus (GAS) is a leading human bacterial pathogen with diverse clinical manifestations. Macrophages constitute a critical first line of host defense against GAS infection, using numerous surface and intracellular receptors such as Toll-like receptors and inflammasomes for pathogen recognition and activation of inflammatory signaling pathways. Depending on the intensity of the GAS infection, activation of these signaling cascades may provide a beneficial early alarm for effective immune clearance, or conversely, may cause hyperinflammation and tissue injury during severe invasive infection. Although traditionally considered an extracellular pathogen, GAS can invade and replicate within macrophages using specific molecular mechanisms to resist phagolysosomal and xenophagic killing. Unraveling GAS-macrophage encounters may reveal new treatment options for this leading agent of infection-associated mortality. [Formula: see text].
Collapse
Affiliation(s)
- J Andrés Valderrama
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|