1
|
Tang D, Khakzad H, Hjortswang E, Malmström L, Ekström S, Happonen L, Malmström J. Streptolysin O accelerates the conversion of plasminogen to plasmin. Nat Commun 2024; 15:10212. [PMID: 39587097 PMCID: PMC11589678 DOI: 10.1038/s41467-024-54173-6] [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: 06/06/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024] Open
Abstract
Group A Streptococcus (GAS) is a human-specific bacterial pathogen that can exploit the plasminogen-plasmin fibrinolysis system to dismantle blood clots and facilitate its spread and survival within the human host. In this study, we use affinity-enrichment mass spectrometry to decipher the host-pathogen protein-protein interaction between plasminogen and streptolysin O, a key cytolytic toxin produced by GAS. This interaction accelerates the conversion of plasminogen to plasmin by both the host tissue-type plasminogen activator and streptokinase, a bacterial plasminogen activator secreted by GAS. Integrative structural mass spectrometry analysis shows that the interaction induces local conformational shifts in plasminogen. These changes lead to the formation of a stabilised intermediate plasminogen-streptolysin O complex that becomes significantly more susceptible to proteolytic processing by plasminogen activators. Our findings reveal a conserved and moonlighting pathomechanistic function for streptolysin O that extends beyond its well-characterised cytolytic activity.
Collapse
Affiliation(s)
- Di Tang
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.
| | - Hamed Khakzad
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, France
| | - Elisabeth Hjortswang
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lars Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Simon Ekström
- SciLifeLab, Integrated Structural Biology Platform, Structural Proteomics Unit Sweden, Lund University, Lund, Sweden
| | - Lotta Happonen
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.
- SciLifeLab, Integrated Structural Biology Platform, Structural Proteomics Unit Sweden, Lund University, Lund, Sweden.
| |
Collapse
|
2
|
Isidoro-Ayza M, Lorch JM, Klein BS. The skin I live in: Pathogenesis of white-nose syndrome of bats. PLoS Pathog 2024; 20:e1012342. [PMID: 39207947 PMCID: PMC11361426 DOI: 10.1371/journal.ppat.1012342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
The emergence of white-nose syndrome (WNS) in North America has resulted in mass mortalities of hibernating bats and total extirpation of local populations. The need to mitigate this disease has stirred a significant body of research to understand its pathogenesis. Pseudogymnoascus destructans, the causative agent of WNS, is a psychrophilic (cold-loving) fungus that resides within the class Leotiomycetes, which contains mainly plant pathogens and is unrelated to other consequential pathogens of animals. In this review, we revisit the unique biology of hibernating bats and P. destructans and provide an updated analysis of the stages and mechanisms of WNS progression. The extreme life history of hibernating bats, the psychrophilic nature of P. destructans, and its evolutionary distance from other well-characterized animal-infecting fungi translate into unique host-pathogen interactions, many of them yet to be discovered.
Collapse
Affiliation(s)
- Marcos Isidoro-Ayza
- Department of Pediatrics, Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jeffrey M. Lorch
- U.S. Geological Survey, National Wildlife Health Center, Madison, Wisconsin, United States of America
| | - Bruce S. Klein
- Department of Pediatrics, Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
3
|
Isidoro-Ayza M, Klein BS. Pathogenic strategies of Pseudogymnoascus destructans during torpor and arousal of hibernating bats. Science 2024; 385:194-200. [PMID: 38991070 DOI: 10.1126/science.adn5606] [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: 12/15/2023] [Accepted: 05/14/2024] [Indexed: 07/13/2024]
Abstract
Millions of hibernating bats across North America have died from white-nose syndrome (WNS), an emerging disease caused by a psychrophilic (cold-loving) fungus, Pseudogymnoascus destructans, that invades their skin. Mechanisms of P. destructans invasion of bat epidermis remain obscure. Guided by our in vivo observations, we modeled hibernation with a newly generated little brown bat (Myotis lucifugus) keratinocyte cell line. We uncovered the stealth intracellular lifestyle of P. destructans, which inhibits apoptosis of keratinocytes and spreads through the cells by two epidermal growth factor receptor (EGFR)-dependent mechanisms: active penetration during torpor and induced endocytosis during arousal. Melanin of endocytosed P. destructans blocks endolysosomal maturation, facilitating P. destructans survival and germination after return to torpor. Blockade of EGFR aborts P. destructans entry into keratinocytes.
Collapse
Affiliation(s)
- Marcos Isidoro-Ayza
- Department of Pediatrics, Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bruce S Klein
- Department of Pediatrics, Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
4
|
Nishioka ST, Snipper J, Lee J, Schapiro J, Zhang RZ, Abe H, Till A, Okumura CYM. Group A Streptococcus induces lysosomal dysfunction in THP-1 macrophages. Infect Immun 2024; 92:e0014124. [PMID: 38722166 PMCID: PMC11237432 DOI: 10.1128/iai.00141-24] [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: 04/01/2024] [Accepted: 04/11/2024] [Indexed: 06/12/2024] Open
Abstract
The human-specific bacterial pathogen group A Streptococcus (GAS) is a significant cause of morbidity and mortality. Macrophages are important to control GAS infection, but previous data indicate that GAS can persist in macrophages. In this study, we detail the molecular mechanisms by which GAS survives in THP-1 macrophages. Our fluorescence microscopy studies demonstrate that GAS is readily phagocytosed by macrophages, but persists within phagolysosomes. These phagolysosomes are not acidified, which is in agreement with our findings that GAS cannot survive in low pH environments. We find that the secreted pore-forming toxin Streptolysin O (SLO) perforates the phagolysosomal membrane, allowing leakage of not only protons but also large proteins including the lysosomal protease cathepsin B. Additionally, GAS recruits CD63/LAMP-3, which may contribute to lysosomal permeabilization, especially in the absence of SLO. Thus, although GAS does not inhibit fusion of the lysosome with the phagosome, it has multiple mechanisms to prevent proper phagolysosome function, allowing for persistence of the bacteria within the macrophage. This has important implications for not only the initial response but also the overall functionality of the macrophages, which may lead to the resulting pathologies in GAS infection. Our data suggest that therapies aimed at improving macrophage function may positively impact patient outcomes in GAS infection.
Collapse
Affiliation(s)
- Scott T Nishioka
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Joshua Snipper
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Jimin Lee
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Joshua Schapiro
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Robert Z Zhang
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Hyewon Abe
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Andreas Till
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
- The San Diego Center for Systems Biology, University of California San Diego, La Jolla, California, USA
- University Hospital of Bonn, Bonn, Germany
| | | |
Collapse
|
5
|
Su MSW, Cheng YL, Lin YS, Wu JJ. Interplay between group A Streptococcus and host innate immune responses. Microbiol Mol Biol Rev 2024; 88:e0005222. [PMID: 38451081 PMCID: PMC10966951 DOI: 10.1128/mmbr.00052-22] [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] [Indexed: 03/08/2024] Open
Abstract
SUMMARYGroup A Streptococcus (GAS), also known as Streptococcus pyogenes, is a clinically well-adapted human pathogen that harbors rich virulence determinants contributing to a broad spectrum of diseases. GAS is capable of invading epithelial, endothelial, and professional phagocytic cells while evading host innate immune responses, including phagocytosis, selective autophagy, light chain 3-associated phagocytosis, and inflammation. However, without a more complete understanding of the different ways invasive GAS infections develop, it is difficult to appreciate how GAS survives and multiplies in host cells that have interactive immune networks. This review article attempts to provide an overview of the behaviors and mechanisms that allow pathogenic GAS to invade cells, along with the strategies that host cells practice to constrain GAS infection. We highlight the counteractions taken by GAS to apply virulence factors such as streptolysin O, nicotinamide-adenine dinucleotidase, and streptococcal pyrogenic exotoxin B as a hindrance to host innate immune responses.
Collapse
Affiliation(s)
- Marcia Shu-Wei Su
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Lin Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yee-Shin Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jiunn-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
6
|
Alamiri F, André O, De S, Nordenfelt P, Hakansson AP. Role of serotype and virulence determinants of Streptococcus pyogenes biofilm bacteria in internalization and persistence in epithelial cells in vitro. Front Cell Infect Microbiol 2023; 13:1146431. [PMID: 37234777 PMCID: PMC10206268 DOI: 10.3389/fcimb.2023.1146431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Streptococcus pyogenes causes a multitude of local and systemic infections, the most common being pharyngitis in children. Recurrent pharyngeal infections are common and are thought to be due to the re-emergence of intracellular GAS upon completion of antibiotic treatment. The role of colonizing biofilm bacteria in this process is not fully clear. Here, live respiratory epithelial cells were inoculated with broth-grown or biofilm bacteria of different M-types, as well as with isogenic mutants lacking common virulence factors. All M-types tested adhered to and were internalized into epithelial cells. Interestingly, internalization and persistence of planktonic bacteria varied significantly between strains, whereas biofilm bacteria were internalized in similar and higher numbers, and all strains persisted beyond 44 hours, showing a more homogenous phenotype. The M3 protein, but not the M1 or M5 proteins, was required for optimal uptake and persistence of both planktonic and biofilm bacteria inside cells. Moreover, the high expression of capsule and SLO inhibited cellular uptake and capsule expression was required for intracellular survival. Streptolysin S was required for optimal uptake and persistence of M3 planktonic bacteria, whereas SpeB improved intracellular survival of biofilm bacteria. Microscopy of internalized bacteria showed that planktonic bacteria were internalized in lower numbers as individual or small clumps of bacteria in the cytoplasm, whereas GAS biofilm bacteria displayed a pattern of perinuclear localization of bacterial aggregates that affected actin structure. Using inhibitors targeting cellular uptake pathways, we confirmed that planktonic GAS mainly uses a clathrin-mediated uptake pathway that also required actin and dynamin. Clathrin was not involved in biofilm internalization, but internalization required actin rearrangement and PI3 kinase activity, possibly suggesting macropinocytosis. Together these results provide a better understanding of the potential mechanisms of uptake and survival of various phenotypes of GAS bacteria relevant for colonization and recurrent infection.
Collapse
Affiliation(s)
- Feiruz Alamiri
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Oscar André
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Supradipta De
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Anders P. Hakansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
7
|
Banerji R, Saroj SD. Exposure to Acyl Homoserine Lactone Enhances Survival of Streptococcus pyogenes in Murine Macrophages. MICROBIAL ECOLOGY 2022; 84:1256-1263. [PMID: 34782937 DOI: 10.1007/s00248-021-01926-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
Streptococcus pyogenes is an opportunistic pathogen causing infections of the skin and upper respiratory tract of the human host. Due to the polymicrobial community present in the human host, S. pyogenes comes across several interspecies signalling molecules. Among these molecules, N-(3-oxododecanoyl)-L-homoserine lactone (Oxo-C12) modulates the morphology, thereby enhancing virulence characteristics of S. pyogenes. After the initial attachment of the bacteria to the host cell, the pathogen needs to invade the host immune system for a successful infection to occur. The host immune system is activated upon infection, where macrophages engulf the pathogen, thereby killing the bacteria. However, S. pyogenes have evolved various strategies to evade the host immune response. In this study, we investigate the role of Oxo-C12 in enhancing the survival of S. pyogenes M3 in murine macrophages. The observed Oxo-C12-mediated increased survival in murine macrophages was through increased lysozyme and acid stress resistance. Moreover, Oxo-C12 increased the survival of S. pyogenes in normal human serum. Thus, understanding the role of interspecies signalling in enhancing the survival strategies of S. pyogenes in the host will further help fill the gap for therapeutics development.
Collapse
Affiliation(s)
- Rajashri Banerji
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Symbiosis Knowledge Village, Lavale, Pune, 412115, Maharashtra, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Symbiosis Knowledge Village, Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
8
|
Banerji R, Iyer P, Saroj SD. Spermidine enhances the survival of Streptococcus pyogenes M3 under oxidative stress. Mol Oral Microbiol 2022; 37:53-62. [PMID: 34994090 DOI: 10.1111/omi.12360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022]
Abstract
Streptococcus pyogenes, a host-restricted gram-positive pathogen during infection, initially adheres to the epithelia of the nasopharynx and respiratory tract of the human host, followed by disseminating to other organs and evading the host immune system. Upon phagocytosis, S. pyogenes encounters oxidative stress inside the macrophages. The role of polyamines in regulating various physiological functions including stress resistance in bacteria has been reported widely. Since S. pyogenes lacks the machinery for the biosynthesis of polyamines, the study aimed to understand the role of extracellular polyamines in the survival of S. pyogenes under oxidative stress environments. S. pyogenes being a catalase-negative organism, we report that its survival within the macrophages and H2 O2 is enhanced by the presence of spermidine. The increased survival can be attributed to the upregulation of oxidative stress response genes such as sodM, npx, and mtsABC. In addition, spermidine influences the upregulation of virulence factors such as sagA, slo, and hasA. Also, spermidine leads to a decrease in hydrophobicity of the cell membrane and an increase in hyaluronic acid. This study suggests a role for extracellular spermidine in the survival of S. pyogenes under oxidative stress environments. Recognizing the factors that modulate S. pyogenes survival and virulence under stress will assist in understanding its interactions with the host.
Collapse
Affiliation(s)
- Rajashri Banerji
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Parvati Iyer
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
| |
Collapse
|
9
|
A Role of Epithelial Cells and Virulence Factors in Biofilm Formation by Streptococcus pyogenes In Vitro. Infect Immun 2020; 88:IAI.00133-20. [PMID: 32661124 DOI: 10.1128/iai.00133-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/07/2020] [Indexed: 02/05/2023] Open
Abstract
Biofilm formation by Streptococcus pyogenes (group A streptococcus [GAS]) in model systems mimicking the respiratory tract is poorly documented. Most studies have been conducted on abiotic surfaces, which poorly represent human tissues. We have previously shown that GAS forms mature and antibiotic-resistant biofilms on physiologically relevant epithelial cells. However, the roles of the substratum, extracellular matrix (ECM) components, and GAS virulence factors in biofilm formation and structure are unclear. In this study, biofilm formation was measured on respiratory epithelial cells and keratinocytes by determining biomass and antibiotic resistance, and biofilm morphology was visualized using scanning electron microscopy. All GAS isolates tested formed biofilms that had similar, albeit not identical, biomass and antibiotic resistance for both cell types. Interestingly, functionally mature biofilms formed more rapidly on keratinocytes but were structurally denser and coated with more ECM on respiratory epithelial cells. The ECM was crucial for biofilm integrity, as protein- and DNA-degrading enzymes induced bacterial release from biofilms. Abiotic surfaces supported biofilm formation, but these biofilms were structurally less dense and organized. No major role for M protein, capsule, or streptolysin O was observed in biofilm formation on epithelial cells, although some morphological differences were detected. NAD-glycohydrolase was required for optimal biofilm formation, whereas streptolysin S and cysteine protease SpeB impaired this process. Finally, no correlation was found between cell adherence or autoaggregation and GAS biofilm formation. Combined, these results provide a better understanding of the role of biofilm formation in GAS pathogenesis and can potentially provide novel targets for future treatments against GAS infections.
Collapse
|
10
|
Matsumura T, Nishiyama A, Aiko M, Ainai A, Ikebe T, Chiba J, Ato M, Takahashi Y. An anti-perfringolysin O monoclonal antibody cross-reactive with streptolysin O protects against streptococcal toxic shock syndrome. BMC Res Notes 2020; 13:419. [PMID: 32891180 PMCID: PMC7487723 DOI: 10.1186/s13104-020-05264-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/01/2020] [Indexed: 11/17/2022] Open
Abstract
Objective Streptococcus pyogenes (Group A Streptococcus; GAS) causes a variety of infections that include life-threatening, severe invasive GAS infections, such as streptococcal toxic shock syndrome (STSS), with > 30% mortality rate, despite effective antibiotics and treatment options. STSS clinical isolates highly express streptolysin O (SLO), a member of a large family of pore-forming toxins called cholesterol-dependent cytolysins (CDCs). SLO is an important toxic factor for GAS and may be an effective therapeutic target for the treatment of STSS. Our aim was to identify a monoclonal antibody (mAb) that reacts with SLO and has therapeutic potential for STSS treatment. Results We focused on mAbs that had originally been established as neutralizing reagents to perfringolysin O (PFO), another member of the CDC family, as some cross-reactivity with SLO had been reported. Here, we confirmed cross-reactivity of an anti-PFO mAb named HS1 with SLO. In vitro analysis revealed that HS1 mAb sufficiently prevented human neutrophils from being killed by STSS clinical isolates. Furthermore, prophylactic and therapeutic injection of HS1 mAb into C57BL/6 mice significantly improved the survival rate following lethal infection with an STSS clinical isolate. These results highlight the therapeutic potential of HS1 mAb for STSS treatment.
Collapse
Affiliation(s)
- Takayuki Matsumura
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| | - Ayae Nishiyama
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Michio Aiko
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Tadayoshi Ikebe
- Department of Bacteriology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Joe Chiba
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Manabu Ato
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashimurayamashi, Tokyo, 189-0002, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| |
Collapse
|
11
|
Vyas HKN, Proctor EJ, McArthur J, Gorman J, Sanderson-Smith M. Current Understanding of Group A Streptococcal Biofilms. Curr Drug Targets 2020; 20:982-993. [PMID: 30947646 PMCID: PMC6700754 DOI: 10.2174/1389450120666190405095712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 02/01/2023]
Abstract
Background: It has been proposed that GAS may form biofilms. Biofilms are microbial communities that aggregate on a surface, and exist within a self-produced matrix of extracellular polymeric substances. Biofilms offer bacteria an increased survival advantage, in which bacteria persist, and resist host immunity and antimicrobial treatment. The biofilm phenotype has long been recognized as a virulence mechanism for many Gram-positive and Gram-negative bacteria, however very little is known about the role of biofilms in GAS pathogenesis. Objective: This review provides an overview of the current knowledge of biofilms in GAS pathogenesis. This review assesses the evidence of GAS biofilm formation, the role of GAS virulence factors in GAS biofilm formation, modelling GAS biofilms, and discusses the polymicrobial nature of biofilms in the oropharynx in relation to GAS. Conclusion: Further study is needed to improve the current understanding of GAS as both a mono-species biofilm, and as a member of a polymicrobial biofilm. Improved modelling of GAS biofilm formation in settings closely mimicking in vivo conditions will ensure that biofilms generated in the lab closely reflect those occurring during clinical infection.
Collapse
Affiliation(s)
- Heema K N Vyas
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Emma-Jayne Proctor
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Jason McArthur
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Jody Gorman
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Martina Sanderson-Smith
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| |
Collapse
|
12
|
Singh S, Yabaji SM, Ali R, Srivastava KK, Haq W. Synthesis and biological activity of Ub2 derived peptides as potential host-directed antitubercular therapy. Chem Biol Drug Des 2019; 94:1330-1338. [PMID: 30805971 DOI: 10.1111/cbdd.13508] [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: 11/18/2018] [Revised: 01/24/2019] [Accepted: 02/09/2019] [Indexed: 11/27/2022]
Abstract
The correlation of mycobactericidal property of macrophages with its potential to deliver bacteria to hydrolytic lysosomes, augmented with ubiquitin-derived peptides (Ub2), activates the process of autophagy. This leads to the formation of phagolysosomes supported by factor involving increased cationic charges which regulate the acidic pH causing elimination of Mycobacterium. To better understand this interaction of cationic-rich ubiquitin-derived peptides with mycobacteria and to identify putative mycobacterial intrinsic resistance mechanisms for phagolysosome formation, we have synthesized a new series of Ub2 peptides, wherein the Gly residues are replaced with azaGly with the aim to improve metabolic stability. In addition to that a new methodology is reported for the synthesis of heteroaryl tethered peptides using azaGly as a linker. We have demonstrated that positive puncta (directly proportional to the acidification of lysosome) in cytosol was significantly increased after 6 hours on the treatment of macrophage with Ub2 peptide derivatives (1, 6, 10, and 11) causing the higher intensity of lysosome observed through LysoTracker Red Dye. The circular dichroism spectral studies are carried out in water and water:TFE mixture and demonstrated that the Ub2 peptides have helix-forming tendency in the presence of TFE. The recognizable intracellular killing of Mycobacterium tuberculosis by Ub2 peptides provides a new approach for host-directed therapy.
Collapse
Affiliation(s)
- Shalini Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shivraj M Yabaji
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rafat Ali
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kishore K Srivastava
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research, New Delhi, India
| | - Wahajul Haq
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
13
|
Vajjala A, Biswas D, Tay WH, Hanski E, Kline KA. Streptolysin-induced endoplasmic reticulum stress promotes group A Streptococcal host-associated biofilm formation and necrotising fasciitis. Cell Microbiol 2018; 21:e12956. [PMID: 30239106 DOI: 10.1111/cmi.12956] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/24/2022]
Abstract
Group A Streptococcus (GAS) is a human pathogen that causes infections ranging from mild to fulminant and life-threatening. Biofilms have been implicated in acute GAS soft-tissue infections such as necrotising fasciitis (NF). However, most in vitro models used to study GAS biofilms have been designed to mimic chronic infections and insufficiently recapitulate in vivo conditions along with the host-pathogen interactions that might influence biofilm formation. Here, we establish and characterise an in vitro model of GAS biofilm development on mammalian cells that simulates microcolony formation observed in a mouse model of human NF. We show that on mammalian cells, GAS forms dense aggregates that display hallmark biofilm characteristics including a 3D architecture and enhanced tolerance to antibiotics. In contrast to abiotic-grown biofilms, host-associated biofilms require the expression of secreted GAS streptolysins O and S (SLO, SLS) that induce endoplasmic reticulum (ER) stress in the host. In an in vivo mouse model, the streptolysin null mutant is attenuated in both microcolony formation and bacterial spread, but pretreatment of soft-tissue with an ER stressor restores the ability of the mutant to form wild-type-like microcolonies that disseminate throughout the soft tissue. Taken together, we have identified a new role of streptolysin-driven ER stress in GAS biofilm formation and NF disease progression.
Collapse
Affiliation(s)
- Anuradha Vajjala
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | - Debabrata Biswas
- Cellular and Molecular Mechanisms of Inflammation, Campus for Research Excellence and Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore (NUS)-The Hebrew University of Jerusalem (HUJ), Singapore
| | - Wei Hong Tay
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Interdisciplinary Graduate School, Nanyang Technological University, Singapore
| | - Emanuel Hanski
- Cellular and Molecular Mechanisms of Inflammation, Campus for Research Excellence and Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore (NUS)-The Hebrew University of Jerusalem (HUJ), Singapore.,Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kimberly A Kline
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
14
|
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
|
15
|
Escajadillo T, Olson J, Luk BT, Zhang L, Nizet V. A Red Blood Cell Membrane-Camouflaged Nanoparticle Counteracts Streptolysin O-Mediated Virulence Phenotypes of Invasive Group A Streptococcus. Front Pharmacol 2017; 8:477. [PMID: 28769806 PMCID: PMC5513932 DOI: 10.3389/fphar.2017.00477] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/03/2017] [Indexed: 12/16/2022] Open
Abstract
Group A Streptococcus (GAS), an important human-specific Gram-positive bacterial pathogen, is associated with a broad spectrum of disease, ranging from mild superficial infections such as pharyngitis and impetigo, to serious invasive infections including necrotizing fasciitis and streptococcal toxic shock syndrome. The GAS pore-forming streptolysin O (SLO) is a well characterized virulence factor produced by nearly all GAS clinical isolates. High level expression of SLO is epidemiologically linked to intercontinental dissemination of hypervirulent clonotypes and poor clinical outcomes. SLO can trigger macrophage and neutrophil cell death and/or the inactivation of immune cell functions, and promotes tissue injury and bacterial survival in animal models of infection. In the present work, we describe how the pharmacological presentation of red blood cell (RBC) derived biomimetic nanoparticles ("nanosponges") can sequester SLO and block the ability of GAS to damage host cells, thereby preserving innate immune function and increasing bacterial clearance in vitro and in vivo. Nanosponge administration protected human neutrophils, macrophages, and keratinocytes against SLO-mediated cytotoxicity. This therapeutic intervention prevented SLO-induced macrophage apoptosis and increased neutrophil extracellular trap formation, allowing increased GAS killing by the respective phagocytic cell types. In a murine model of GAS necrotizing skin infection, local administration of the biomimetic nanosponges was associated with decreased lesion size and reduced bacterial colony-forming unit recovery. Utilization of a toxin decoy and capture platform that inactivates the secreted SLO before it contacts the host cell membrane, presents a novel virulence factor targeted strategy that could be a powerful adjunctive therapy in severe GAS infections where morbidity and mortality are high despite antibiotic treatment.
Collapse
Affiliation(s)
- Tamara Escajadillo
- Biomedical Sciences Graduate Program, University of California, San Diego, La JollaCA, United States.,Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La JollaCA, United States
| | - Joshua Olson
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La JollaCA, United States
| | - Brian T Luk
- Department of NanoEngineering, University of California, San Diego, La JollaCA, United States
| | - Liangfang Zhang
- Department of NanoEngineering, University of California, San Diego, La JollaCA, United States
| | - Victor Nizet
- Biomedical Sciences Graduate Program, University of California, San Diego, La JollaCA, United States.,Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La JollaCA, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La JollaCA, United States
| |
Collapse
|
16
|
Zou X, Pan T, Chen L, Tian Y, Zhang W. Luminescence materials for pH and oxygen sensing in microbial cells - structures, optical properties, and biological applications. Crit Rev Biotechnol 2016; 37:723-738. [PMID: 27627832 DOI: 10.1080/07388551.2016.1223011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Luminescence including fluorescence and phosphorescence sensors have been demonstrated to be important for studying cell metabolism, and diagnosing diseases and cancer. Various design principles have been employed for the development of sensors in different formats, such as organic molecules, polymers, polymeric hydrogels, and nanoparticles. The integration of the sensing with fluorescence imaging provides valuable tools for biomedical research and applications at not only bulk-cell level but also at single-cell level. In this article, we critically reviewed recent progresses on pH, oxygen, and dual pH and oxygen sensors specifically for their application in microbial cells. In addition, we focused not only on sensor materials with different chemical structures, but also on design and applications of sensors for better understanding cellular metabolism of microbial cells. Finally, we also provided an outlook for future materials design and key challenges in reaching broad applications in microbial cells.
Collapse
Affiliation(s)
- Xianshao Zou
- a Department of Materials Science and Engineering , South University of Science and Technology of China , Shenzhen , Guangdong , P.R. China
| | - Tingting Pan
- a Department of Materials Science and Engineering , South University of Science and Technology of China , Shenzhen , Guangdong , P.R. China
| | - Lei Chen
- b Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology , Tianjin University , Tianjin , P.R. China.,c Key Laboratory of Systems Bioengineering, Ministry of Education of China , Tianjin , P.R. China.,d SynBio Platform, Collaborative Innovation Center of Chemical Science and Engineering , Tianjin , P.R. China
| | - Yanqing Tian
- a Department of Materials Science and Engineering , South University of Science and Technology of China , Shenzhen , Guangdong , P.R. China
| | - Weiwen Zhang
- b Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology , Tianjin University , Tianjin , P.R. China.,c Key Laboratory of Systems Bioengineering, Ministry of Education of China , Tianjin , P.R. China.,d SynBio Platform, Collaborative Innovation Center of Chemical Science and Engineering , Tianjin , P.R. China
| |
Collapse
|
17
|
Abstract
UNLABELLED As key components of innate immune defense, macrophages are essential in controlling bacterial pathogens, including group A Streptococcus(GAS). Despite this, only a limited number of studies have analyzed the recovery of GAS from within human neutrophils and macrophages. Here, we determined the intracellular fate of GAS in human macrophages by using several quantitative approaches. In both U937 and primary human macrophages, the appearance over time of long GAS chains revealed that despite GAS-mediated cytotoxicity, replication occurred in viable, propidium iodide-negative macrophages. Whereas the major virulence factor M1 did not contribute to bacterial growth, a GAS mutant strain deficient in streptolysin O (SLO) was impaired for intracellular replication. SLO promoted bacterial escape from the GAS-containing vacuole (GCV) into the macrophage cytosol. Up to half of the cytosolic GAS colocalized with ubiquitin and p62, suggesting that the bacteria were targeted by the autophagy machinery. Despite this, live imaging of U937 macrophages revealed proficient replication of GAS after GCV rupture, indicating that escape from the GCV is important for growth of GAS in macrophages. Our results reveal that GAS can replicate within viable human macrophages, with SLO promoting GCV escape and cytosolic growth, despite the recruitment of autophagy receptors to bacteria. IMPORTANCE Classically regarded as an extracellular pathogen, GAS can persist within human epithelial cells, as well as neutrophils and macrophages. Some studies suggest that GAS can modulate its intracellular vacuole to promote survival and perhaps replicate in macrophages. However, an in-depth single-cell analysis of the dynamics of survival and replication is lacking. We used macrophage-like cell lines and primary macrophages to measure the intracellular growth of GAS at both the population and single-cell levels. While CFU counts revealed no increase in overall bacterial growth, quantitative fluorescence microscopy, flow cytometry, and time-lapse imaging revealed bacterial replication in a proportion of infected macrophages. This study emphasizes the importance of single-cell analysis especially when studying the intracellular fate of a pathogen that is cytotoxic and displays heterogeneity in terms of intracellular killing and growth. To our knowledge, this study provides the first direct visualization of GAS replication inside human cells.
Collapse
|
18
|
NAD+-Glycohydrolase Promotes Intracellular Survival of Group A Streptococcus. PLoS Pathog 2016; 12:e1005468. [PMID: 26938870 PMCID: PMC4777570 DOI: 10.1371/journal.ppat.1005468] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/02/2016] [Indexed: 01/19/2023] Open
Abstract
A global increase in invasive infections due to group A Streptococcus (S. pyogenes or GAS) has been observed since the 1980s, associated with emergence of a clonal group of strains of the M1T1 serotype. Among other virulence attributes, the M1T1 clone secretes NAD+-glycohydrolase (NADase). When GAS binds to epithelial cells in vitro, NADase is translocated into the cytosol in a process mediated by streptolysin O (SLO), and expression of these two toxins is associated with enhanced GAS intracellular survival. Because SLO is required for NADase translocation, it has been difficult to distinguish pathogenic effects of NADase from those of SLO. To resolve the effects of the two proteins, we made use of anthrax toxin as an alternative means to deliver NADase to host cells, independently of SLO. We developed a novel method for purification of enzymatically active NADase fused to an amino-terminal fragment of anthrax toxin lethal factor (LFn-NADase) that exploits the avid, reversible binding of NADase to its endogenous inhibitor. LFn-NADase was translocated across a synthetic lipid bilayer in vitro in the presence of anthrax toxin protective antigen in a pH-dependent manner. Exposure of human oropharyngeal keratinocytes to LFn-NADase in the presence of protective antigen resulted in cytosolic delivery of NADase activity, inhibition of protein synthesis, and cell death, whereas a similar construct of an enzymatically inactive point mutant had no effect. Anthrax toxin-mediated delivery of NADase in an amount comparable to that observed during in vitro infection with live GAS rescued the defective intracellular survival of NADase-deficient GAS and increased the survival of SLO-deficient GAS. Confocal microscopy demonstrated that delivery of LFn-NADase prevented intracellular trafficking of NADase-deficient GAS to lysosomes. We conclude that NADase mediates cytotoxicity and promotes intracellular survival of GAS in host cells. Invasive infections due to group A Streptococcus (S. pyogenes or GAS) have become more frequent since the 1980s due, in part, to the emergence and global spread of closely related strains of the M1T1 serotype. A feature of this clonal group is the production of a secreted enzyme, NAD+-glycohydrolase (NADase), which has been suggested to contribute to GAS virulence by intoxication of host cells. For NADase to exert its toxic effects, it must be translocated into the host cell by a second GAS protein, streptolysin O (SLO). SLO is a pore-forming toxin that damages cell membranes in addition to its role in translocating NADase. In order to distinguish effects of NADase on host cell biology from those of SLO, we used components of anthrax toxin to deliver NADase to human throat epithelial cells, independently of SLO. Introduction of NADase into GAS-infected cells increased the intracellular survival of GAS lacking NADase or SLO, and the increase in bacterial survival correlated with inhibition of intracellular trafficking of GAS to lysosomes that mediate bacterial killing. The results support an important role for NADase in enhancing GAS survival in human epithelial cells, a phenomenon that may contribute to GAS colonization and disease.
Collapse
|
19
|
Lood R, Waldetoft KW, Nordenfelt P. Localization-triggered bacterial pathogenesis. Future Microbiol 2015; 10:1659-68. [PMID: 26437846 DOI: 10.2217/fmb.15.89] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bacterial infections are becoming an increasing problem worldwide and there is a need for a deeper understanding of how bacteria turn pathogenic. Here, we suggest that one answer may be found by taking into account the localization of the bacteria, both at an anatomical level and at a microenvironment level. Both commensals and traditional pathogens alter their interaction with the human host depending on the local surroundings--turning either more or less virulent. These localization effects could derive from the characteristics of different anatomical sites but also from local differences within a microenvironment. In order to understand the adaptive functions of bacterial virulence factors, we need to study the bacteria in the environments where they have evolved.
Collapse
Affiliation(s)
- Rolf Lood
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | | | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| |
Collapse
|
20
|
Perobelli SM, Galvani RG, Gonçalves-Silva T, Xavier CR, Nóbrega A, Bonomo A. Plasticity of neutrophils reveals modulatory capacity. ACTA ACUST UNITED AC 2015; 48:665-75. [PMID: 26108096 PMCID: PMC4541684 DOI: 10.1590/1414-431x20154524] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/17/2015] [Indexed: 12/22/2022]
Abstract
Neutrophils are widely known as proinflammatory cells associated with tissue damage
and for their early arrival at sites of infection, where they exert their phagocytic
activity, release their granule contents, and subsequently die. However, this view
has been challenged by emerging evidence that neutrophils have other activities and
are not so short-lived. Following activation, neutrophil effector functions include
production and release of granule contents, reactive oxygen species (ROS), and
neutrophil extracellular traps (NETs). Neutrophils have also been shown to produce a
wide range of cytokines that have pro- or anti-inflammatory activity, adding a
modulatory role for this cell, previously known as a suicide effector. The presence
of cytokines almost always implies intercellular modulation, potentially unmasking
interactions of neutrophils with other immune cells. In fact, neutrophils have been
found to help B cells and to modulate dendritic cell (DC), macrophage, and T-cell
activities. In this review, we describe some ways in which neutrophils influence the
inflammatory environment in infection, cancer, and autoimmunity, regulating both
innate and adaptive immune responses. These cells can switch phenotypes and exert
functions beyond cytotoxicity against invading pathogens, extending the view of
neutrophils beyond suicide effectors to include functions as regulatory and
suppressor cells.
Collapse
Affiliation(s)
- S M Perobelli
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - R G Galvani
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - T Gonçalves-Silva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - C R Xavier
- Departamento de Ciência da Computação, Universidade Federal de São João Del Rei, São João Del Rei, MG, Brasil
| | - A Nóbrega
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - A Bonomo
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
21
|
Latvala S, Mäkelä SM, Miettinen M, Charpentier E, Julkunen I. Dynamin inhibition interferes with inflammasome activation and cytokine gene expression in Streptococcus pyogenes-infected human macrophages. Clin Exp Immunol 2014; 178:320-33. [PMID: 25079511 DOI: 10.1111/cei.12425] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2014] [Indexed: 12/21/2022] Open
Abstract
In the present study, we have analysed the ability of Streptococcus pyogenes [Group A streptococcus (GAS)] to activate the NACHT-domain-, leucine-rich repeat- and PYD-containing protein 3 (NALP3) inflammasome complex in human monocyte-derived macrophages and the molecules and signalling pathways involved in GAS-induced inflammatory responses. We focused upon analysing the impact of dynamin-dependent endocytosis and the role of major streptococcal virulence factors streptolysin O (SLO) and streptolysin S (SLS) in the immune responses induced by GAS. These virulence factors are involved in immune evasion by forming pores in host cell membranes, and aid the bacteria to escape from the endosome-lysosome pathway. We analysed cytokine gene expression in human primary macrophages after stimulation with live or inactivated wild-type GAS as well as with live SLO and SLS defective bacteria. Interleukin (IL)-1β, IL-10, tumour necrosis factor (TNF)-α and chemokine (C-X-C motif) ligand (CXCL)-10 cytokines were produced after bacterial stimulation in a dose-dependent manner and no differences in cytokine levels were seen between live, inactivated or mutant bacteria. These data suggest that streptolysins or other secreted bacterial products are not required for the inflammatory responses induced by GAS. Our data indicate that inhibition of dynamin-dependent endocytosis in macrophages attenuates the induction of IL-1β, TNF-α, interferon (IFN)-β and CXCL-10 mRNAs. We also observed that pro-IL-1β protein was expressed and efficiently cleaved into mature-IL-1β via inflammasome activation after bacterial stimulation. Furthermore, we demonstrate that multiple signalling pathways are involved in GAS-stimulated inflammatory responses in human macrophages.
Collapse
Affiliation(s)
- S Latvala
- Virology Unit, Department of Infectious Disease Surveillance and Control, National Institute for Health and Welfare, Helsinki, Finland
| | | | | | | | | |
Collapse
|
22
|
A MyD88-JAK1-STAT1 complex directly induces SOCS-1 expression in macrophages infected with Group A Streptococcus. Cell Mol Immunol 2014; 12:373-83. [PMID: 25399770 DOI: 10.1038/cmi.2014.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022] Open
Abstract
Some pathogens can use host suppressor of cytokine signaling 1 (SOCS-1), an important negative-feedback molecule, as the main mode of immune evasion. Here we found that group A Streptococcus (GAS) is capable of inducing SOCS-1 expression in RAW264.7 and BMDM macrophages. IFN-β plays a role in GAS-induced SOCS-1 expression in macrophages following the induction of cytokine expression by GAS, representing the classical pathway of SOCS-1 expression. However, GAS also induced STAT1 activation and SOCS-1 expression when GAS-infected cells were incubated with anti-IFN-β monoclonal antibody in this study. Moreover, upon comparing TLR4(-/-) BMDM macrophages with wild-type (WT) cells, we found that TLR4 also plays an essential role in the induction of SOCS-1. MyD88, which is an adaptor protein for TLR4, contributes to STAT1 activation and phosphorylation by forming a complex with Janus kinase 1 (JAK1) and signal transducer and activator of transcription 1 (STAT1) in macrophages. GAS-stimulated expression of STAT1 was severely impaired in MyD88(-/-) macrophages, whereas expression of JAK1 was unaffected, suggesting that MyD88 was involved in STAT1 expression and phosphorylation. Together, these data demonstrated that in addition to IFN-β signaling and MyD88 complex formation, JAK1 and STAT1 act in a novel pathway to directly induce SOCS-1 expression in GAS-infected macrophages, which may be more conducive to rapid bacterial infection.
Collapse
|
23
|
Fieber C, Kovarik P. Responses of innate immune cells to group A Streptococcus. Front Cell Infect Microbiol 2014; 4:140. [PMID: 25325020 PMCID: PMC4183118 DOI: 10.3389/fcimb.2014.00140] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 09/17/2014] [Indexed: 12/22/2022] Open
Abstract
Group A Streptococcus (GAS), also called Streptococcus pyogenes, is a Gram-positive beta-hemolytic human pathogen which causes a wide range of mostly self-limiting but also several life-threatening diseases. Innate immune responses are fundamental for defense against GAS, yet their activation by pattern recognition receptors (PRRs) and GAS-derived pathogen-associated molecular patterns (PAMPs) is incompletely understood. In recent years, the use of animal models together with the powerful tools of human molecular genetics began shedding light onto the molecular mechanisms of innate immune defense against GAS. The signaling adaptor MyD88 was found to play a key role in launching the immune response against GAS in both humans and mice, suggesting that PRRs of the Toll-like receptor (TLR) family are involved in sensing this pathogen. The specific TLRs and their ligands have yet to be identified. Following GAS recognition, induction of cytokines such as TNF and type I interferons (IFNs), leukocyte recruitment, phagocytosis, and the formation of neutrophil extracellular traps (NETs) have been recognized as key events in host defense. A comprehensive knowledge of these mechanisms is needed in order to understand their frequent failure against GAS immune evasion strategies.
Collapse
Affiliation(s)
| | - Pavel Kovarik
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of ViennaVienna, Austria
| |
Collapse
|
24
|
Streptolysin O and NAD-glycohydrolase prevent phagolysosome acidification and promote group A Streptococcus survival in macrophages. mBio 2014; 5:e01690-14. [PMID: 25227466 PMCID: PMC4172074 DOI: 10.1128/mbio.01690-14] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Group A Streptococcus (GAS, Streptococcus pyogenes) is an ongoing threat to human health as the agent of streptococcal pharyngitis, skin and soft tissue infections, and life-threatening conditions such as necrotizing fasciitis and streptococcal toxic shock syndrome. In animal models of infection, macrophages have been shown to contribute to host defense against GAS infection. However, as GAS can resist killing by macrophages in vitro and induce macrophage cell death, it has been suggested that GAS intracellular survival in macrophages may enable persistent infection. Using isogenic mutants, we now show that the GAS pore-forming toxin streptolysin O (SLO) and its cotoxin NAD-glycohydrolase (NADase) mediate GAS intracellular survival and cytotoxicity for macrophages. Unexpectedly, the two toxins did not inhibit fusion of GAS-containing phagosomes with lysosomes but rather prevented phagolysosome acidification. SLO served two essential functions, poration of the phagolysosomal membrane and translocation of NADase into the macrophage cytosol, both of which were necessary for maximal GAS intracellular survival. Whereas NADase delivery to epithelial cells is mediated by SLO secreted from GAS bound to the cell surface, in macrophages, the source of SLO and NADase is GAS contained within phagolysosomes. We found that transfer of NADase from the phagolysosome to the macrophage cytosol occurs not by simple diffusion through SLO pores but rather by a specific translocation mechanism that requires the N-terminal translocation domain of NADase. These results illuminate the mechanisms through which SLO and NADase enable GAS to defeat macrophage-mediated killing and provide new insight into the virulence of a major human pathogen. IMPORTANCE Macrophages constitute an important element of the innate immune response to mucosal pathogens. They ingest and kill microbes by phagocytosis and secrete inflammatory cytokines to recruit and activate other effector cells. Group A Streptococcus (GAS, Streptococcus pyogenes), an important cause of pharyngitis and invasive infections, has been shown to resist killing by macrophages. We find that GAS resistance to macrophage killing depends on the GAS pore-forming toxin streptolysin O (SLO) and its cotoxin NAD-glycohydrolase (NADase). GAS bacteria are internalized by macrophage phagocytosis but resist killing by secreting SLO, which damages the phagolysosome membrane, prevents phagolysosome acidification, and translocates NADase from the phagolysosome into the macrophage cytosol. NADase augments SLO-mediated cytotoxicity by depleting cellular energy stores. These findings may explain the nearly universal production of SLO by GAS clinical isolates and the association of NADase with the global spread of a GAS clone implicated in invasive infections.
Collapse
|
25
|
Okumura CYM, Nizet V. Subterfuge and sabotage: evasion of host innate defenses by invasive gram-positive bacterial pathogens. Annu Rev Microbiol 2014; 68:439-58. [PMID: 25002085 DOI: 10.1146/annurev-micro-092412-155711] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of a severe invasive bacterial infection in an otherwise healthy individual is one of the most striking and fascinating aspects of human medicine. A small cadre of gram-positive pathogens of the genera Streptococcus and Staphylococcus stand out for their unique invasive disease potential and sophisticated ability to counteract the multifaceted components of human innate defense. This review illustrates how these leading human disease agents evade host complement deposition and activation, impede phagocyte recruitment and activation, resist the microbicidal activities of host antimicrobial peptides and reactive oxygen species, escape neutrophil extracellular traps, and promote and accelerate phagocyte cell death through the action of pore-forming cytolysins. Understanding the molecular basis of bacterial innate immune resistance can open new avenues for therapeutic intervention geared to disabling specific virulence factors and resensitizing the pathogen to host innate immune clearance.
Collapse
Affiliation(s)
- Cheryl Y M Okumura
- Department of Biology, Occidental College, Los Angeles, California 90041;
| | | |
Collapse
|
26
|
Disease manifestations and pathogenic mechanisms of Group A Streptococcus. Clin Microbiol Rev 2014. [PMID: 24696436 DOI: 10.1128/cmr.00101-13)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Streptococcus pyogenes, also known as group A Streptococcus (GAS), causes mild human infections such as pharyngitis and impetigo and serious infections such as necrotizing fasciitis and streptococcal toxic shock syndrome. Furthermore, repeated GAS infections may trigger autoimmune diseases, including acute poststreptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease. Combined, these diseases account for over half a million deaths per year globally. Genomic and molecular analyses have now characterized a large number of GAS virulence determinants, many of which exhibit overlap and redundancy in the processes of adhesion and colonization, innate immune resistance, and the capacity to facilitate tissue barrier degradation and spread within the human host. This improved understanding of the contribution of individual virulence determinants to the disease process has led to the formulation of models of GAS disease progression, which may lead to better treatment and intervention strategies. While GAS remains sensitive to all penicillins and cephalosporins, rising resistance to other antibiotics used in disease treatment is an increasing worldwide concern. Several GAS vaccine formulations that elicit protective immunity in animal models have shown promise in nonhuman primate and early-stage human trials. The development of a safe and efficacious commercial human vaccine for the prophylaxis of GAS disease remains a high priority.
Collapse
|
27
|
Walker MJ, Barnett TC, McArthur JD, Cole JN, Gillen CM, Henningham A, Sriprakash KS, Sanderson-Smith ML, Nizet V. Disease manifestations and pathogenic mechanisms of Group A Streptococcus. Clin Microbiol Rev 2014; 27:264-301. [PMID: 24696436 PMCID: PMC3993104 DOI: 10.1128/cmr.00101-13] [Citation(s) in RCA: 582] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Streptococcus pyogenes, also known as group A Streptococcus (GAS), causes mild human infections such as pharyngitis and impetigo and serious infections such as necrotizing fasciitis and streptococcal toxic shock syndrome. Furthermore, repeated GAS infections may trigger autoimmune diseases, including acute poststreptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease. Combined, these diseases account for over half a million deaths per year globally. Genomic and molecular analyses have now characterized a large number of GAS virulence determinants, many of which exhibit overlap and redundancy in the processes of adhesion and colonization, innate immune resistance, and the capacity to facilitate tissue barrier degradation and spread within the human host. This improved understanding of the contribution of individual virulence determinants to the disease process has led to the formulation of models of GAS disease progression, which may lead to better treatment and intervention strategies. While GAS remains sensitive to all penicillins and cephalosporins, rising resistance to other antibiotics used in disease treatment is an increasing worldwide concern. Several GAS vaccine formulations that elicit protective immunity in animal models have shown promise in nonhuman primate and early-stage human trials. The development of a safe and efficacious commercial human vaccine for the prophylaxis of GAS disease remains a high priority.
Collapse
Affiliation(s)
- Mark J. Walker
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Timothy C. Barnett
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Jason D. McArthur
- School of Biological Sciences and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Jason N. Cole
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Christine M. Gillen
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Anna Henningham
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - K. S. Sriprakash
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, Australia
| | - Martina L. Sanderson-Smith
- School of Biological Sciences and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
- Rady Children's Hospital, San Diego, California, USA
| |
Collapse
|
28
|
Feil SC, Ascher DB, Kuiper MJ, Tweten RK, Parker MW. Structural studies of Streptococcus pyogenes streptolysin O provide insights into the early steps of membrane penetration. J Mol Biol 2014; 426:785-92. [PMID: 24316049 PMCID: PMC4323271 DOI: 10.1016/j.jmb.2013.11.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/11/2013] [Accepted: 11/22/2013] [Indexed: 12/22/2022]
Abstract
Cholesterol-dependent cytolysins (CDCs) are a large family of bacterial toxins that exhibit a dependence on the presence of membrane cholesterol in forming large pores in cell membranes. Significant changes in the three-dimensional structure of these toxins are necessary to convert the soluble monomeric protein into a membrane pore. We have determined the crystal structure of the archetypical member of the CDC family, streptolysin O (SLO), a virulence factor from Streptococcus pyogenes. The overall fold is similar to previously reported CDC structures, although the C-terminal domain is in a different orientation with respect to the rest of the molecule. Surprisingly, a signature stretch of CDC sequence called the undecapeptide motif, a key region involved in membrane recognition, adopts a very different structure in SLO to that of the well-characterized CDC perfringolysin O (PFO), although the sequences in this region are identical. An analysis reveals that, in PFO, there are complementary interactions between the motif and the rest of domain 4 that are lost in SLO. Molecular dynamics simulations suggest that the loss of a salt bridge in SLO and a cation-pi interaction are determining factors in the extended conformation of the motif, which in turn appears to result in a greater flexibility of the neighboring L1 loop that houses a cholesterol-sensing motif. These differences may explain the differing abilities of SLO and PFO to efficiently penetrate target cell membranes in the first step of toxin insertion into the membrane.
Collapse
Affiliation(s)
- Susanne C Feil
- ACRF Rational Drug Discovery Centre, Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - David B Ascher
- ACRF Rational Drug Discovery Centre, Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Michael J Kuiper
- Victorian Life Sciences Computation Initiative, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma, Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
29
|
Marks LR, Mashburn-Warren L, Federle MJ, Hakansson AP. Streptococcus pyogenes biofilm growth in vitro and in vivo and its role in colonization, virulence, and genetic exchange. J Infect Dis 2014; 210:25-34. [PMID: 24465015 DOI: 10.1093/infdis/jiu058] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Group A streptococcus (GAS) commonly colonizes the oropharynx and nonintact skin. However, colonization has been little studied and the role of biofilm formation is unclear, as biofilm experiments to date have not been conducted under conditions that mimic the host environment. METHODS In this study we grew GAS biofilms on human keratinocytes under various environmental conditions and used this model to evaluate colonization, invasive disease and natural transformation. RESULTS GAS grown on epithelial cells, but not biofilms grown on abiotic surfaces, produced biofilms with characteristics similar to in vivo colonization. These biofilm bacteria showed a 100-fold higher bacterial burden of nasal-associated lymphoid tissue in mice than broth-grown bacteria, and were not virulent during septic infection, which was attributed in part to down-regulation of genes typically involved in localized and invasive disease. We also showed for the first time that GAS were naturally transformable when grown in biofilms and during colonization of NALT in vivo. CONCLUSIONS These findings provide novel model systems to study biofilm formation of GAS in vitro and in vivo, suggest an important role for biofilm formation during GAS colonization, and provide an explanation for the known genome diversity within the GAS population.
Collapse
|
30
|
Streptolysin O and its co-toxin NAD-glycohydrolase protect group A Streptococcus from Xenophagic killing. PLoS Pathog 2013; 9:e1003394. [PMID: 23762025 PMCID: PMC3675196 DOI: 10.1371/journal.ppat.1003394] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 04/17/2013] [Indexed: 01/01/2023] Open
Abstract
Group A Streptococcus (Streptococcus pyogenes or GAS) causes pharyngitis, severe invasive infections, and the post-infectious syndromes of glomerulonephritis and rheumatic fever. GAS can be internalized and killed by epithelial cells in vitro, a process that may contribute to local innate defense against pharyngeal infection. Secretion of the pore-forming toxin streptolysin O (SLO) by GAS has been reported to stimulate targeted autophagy (xenophagy) upon internalization of the bacteria by epithelial cells. Whereas this process was associated with killing of GAS in HeLa cells, studies in human keratinocytes found SLO production enhanced intracellular survival. To reconcile these conflicting observations, we now report in-depth investigation of xenophagy in response to GAS infection of human oropharyngeal keratinocytes, the predominant cell type of the pharyngeal epithelium. We found that SLO expression was associated with prolonged intracellular survival; unexpectedly, expression of the co-toxin NADase was required for this effect. Enhanced intracellular survival was lost upon deletion of NADase or inactivation of its enzymatic activity. Shortly after internalization of GAS by keratinocytes, SLO-mediated damage to the bacteria-containing vacuole resulted in exposure to the cytosol, ubiquitination of GAS and/or associated vacuolar membrane remnants, and engulfment of GAS in LC3-positive vacuoles. We also found that production of streptolysin S could mediate targeting of GAS to autophagosomes in the absence of SLO, a process accompanied by galectin 8 binding to damaged GAS-containing endosomes. Maturation of GAS-containing autophagosome-like vacuoles to degradative autolysosomes was prevented by SLO pore-formation and by SLO-mediated translocation of enzymatically active NADase into the keratinocyte cytosol. We conclude that SLO stimulates xenophagy in pharyngeal keratinocytes, but the coordinated action of SLO and NADase prevent maturation of GAS-containing autophagosomes, thereby prolonging GAS intracellular survival. This novel activity of NADase to block autophagic killing of GAS in pharyngeal cells may contribute to pharyngitis treatment failure, relapse, and chronic carriage. Group A Streptococcus (Streptococcus pyogenes or GAS) is the agent of streptococcal pharyngitis (strep throat), invasive infections such as necrotizing fasciitis and streptococcal toxic shock, and post-infectious complications including rheumatic heart disease. Epithelial cells internalize and kill GAS in vitro and may contribute to local innate immune defense in the human pharynx. We now find that production of the secreted pore-forming toxin streptolysin O (SLO) triggered targeted autophagy (termed xenophagy) of GAS in human oropharyngeal keratinocytes, but also enhanced GAS intracellular survival. Increased GAS survival was dependent both on pore-formation by SLO and on SLO-mediated translocation of an enzymatically active co-toxin, NAD-glycohydrolase, into the keratinocyte cytosol. The survival-enhancing effect of both toxins was associated with inhibition of lysosomal fusion with GAS-containing autophagosomes to form functional degradative autolysosomes. These findings reveal a novel coordinated role of two streptococcal toxins in protecting GAS from xenophagic killing and enhancing intracellular survival. Prolonged GAS intracellular survival may contribute to pharyngitis treatment failure, relapse, and chronic carriage.
Collapse
|
31
|
Vitamin D and the human antimicrobial peptide LL-37 enhance group a streptococcus resistance to killing by human cells. mBio 2012; 3:mBio.00394-12. [PMID: 23093388 PMCID: PMC3482505 DOI: 10.1128/mbio.00394-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The CsrRS two-component regulatory system of group A Streptococcus (GAS; Streptococcus pyogenes) responds to subinhibitory concentrations of the human antimicrobial peptide LL-37. LL-37 signaling through CsrRS results in upregulation of genes that direct synthesis of virulence factors, including the hyaluronic acid capsule and streptolysin O (SLO). Here, we demonstrate that a consequence of this response is augmented GAS resistance to killing by human oropharyngeal keratinocytes, neutrophils, and macrophages. LL-37-induced upregulation of SLO and hyaluronic acid capsule significantly reduced internalization of GAS by keratinocytes and phagocytic killing by neutrophils and macrophages. Because vitamin D induces LL-37 production by macrophages, we tested its effect on macrophage killing of GAS. In contrast to the reported enhancement of macrophage function in relation to other pathogens, treatment of macrophages with 1α,25-dihydroxy-vitamin D3 paradoxically reduced the ability of macrophages to control GAS infection. These observations demonstrate that LL-37 signals through CsrRS to induce a virulence phenotype in GAS characterized by heightened resistance to ingestion and killing by both epithelial cells and phagocytes. By inducing LL-37 production in macrophages, vitamin D may contribute to this paradoxical exacerbation of GAS infection. IMPORTANCE It remains poorly understood why group A Streptococcus (GAS) causes asymptomatic colonization or localized throat inflammation in most individuals but rarely progresses to invasive infection. The human antimicrobial peptide LL-37, which is produced as part of the innate immune response to GAS infection, signals through the GAS CsrRS two-component regulatory system to upregulate expression of multiple virulence factors. This study reports that two CsrRS-regulated GAS virulence factors-streptolysin O and the hyaluronic acid capsule-are critical in LL-37-induced resistance of GAS to killing by human throat epithelial cells and by neutrophils and macrophages. Vitamin D, which increases LL-37 production in macrophages, has the paradoxical effect of increasing GAS resistance to macrophage-mediated killing. In this way, the human innate immune response may promote the transition from GAS colonization to invasive infection.
Collapse
|
32
|
Fraunholz M, Sinha B. Intracellular Staphylococcus aureus: live-in and let die. Front Cell Infect Microbiol 2012; 2:43. [PMID: 22919634 PMCID: PMC3417557 DOI: 10.3389/fcimb.2012.00043] [Citation(s) in RCA: 275] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 03/15/2012] [Indexed: 12/30/2022] Open
Abstract
Staphylococcus aureus uses a plethora of virulence factors to accommodate a diversity of niches in its human host. Aside from the classical manifestations of S. aureus-induced diseases, the pathogen also invades and survives within mammalian host cells.The survival strategies of the pathogen are as diverse as strains or host cell types used. S. aureus is able to replicate in the phagosome or freely in the cytoplasm of its host cells. It escapes the phagosome of professional and non-professional phagocytes, subverts autophagy, induces cell death mechanisms such as apoptosis and pyronecrosis, and even can induce anti-apoptotic programs in phagocytes. The focus of this review is to present a guide to recent research outlining the variety of intracellular fates of S. aureus.
Collapse
Affiliation(s)
- Martin Fraunholz
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany.
| | | |
Collapse
|
33
|
Abstract
Streptococcus pyogenes is also known as group A Streptococcus (GAS) and is an important human pathogen that causes considerable morbidity and mortality worldwide. The GAS serotype M1T1 clone is the most frequently isolated serotype from life-threatening invasive (at a sterile site) infections, such as streptococcal toxic shock-like syndrome and necrotizing fasciitis. Here, we describe the virulence factors and newly discovered molecular events that mediate the in vivo changes from non-invasive GAS serotype M1T1 to the invasive phenotype, and review the invasive-disease trigger for non-M1 GAS. Understanding the molecular basis and mechanism of initiation for streptococcal invasive disease may expedite the discovery of novel therapeutic targets for the treatment and control of severe invasive GAS diseases.
Collapse
|
34
|
Zinkernagel AS, Hruz P, Uchiyama S, von Köckritz-Blickwede M, Schuepbach RA, Hayashi T, Carson DA, Nizet V. Importance of Toll-like receptor 9 in host defense against M1T1 group A Streptococcus infections. J Innate Immun 2011; 4:213-8. [PMID: 21860217 DOI: 10.1159/000329550] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 05/24/2011] [Indexed: 11/19/2022] Open
Abstract
Timely recognition and elimination of invasive group A Streptococcus (GAS) by innate immunity is crucial to control infection. The intracellular pattern recognition receptor Toll-like receptor 9 (TLR9) promotes macrophage hypoxia-inducible factor-1α levels, oxidative burst and nitric oxide production in response to GAS. TLR9 contributes to GAS clearance in vivo in both localized cutaneous and systemic infection models.
Collapse
Affiliation(s)
- Annelies S Zinkernagel
- Division of Pharmacology and Drug Discovery, Department of Pediatrics, University of California, San Diego, Calif., USA. annelies.zinkernagel @ usz.ch
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bergsbaken T, Fink SL, den Hartigh AB, Loomis WP, Cookson BT. Coordinated host responses during pyroptosis: caspase-1-dependent lysosome exocytosis and inflammatory cytokine maturation. THE JOURNAL OF IMMUNOLOGY 2011; 187:2748-54. [PMID: 21804020 DOI: 10.4049/jimmunol.1100477] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Activation of caspase-1 leads to pyroptosis, a program of cell death characterized by cell lysis and inflammatory cytokine release. Caspase-1 activation triggered by multiple nucleotide-binding oligomerization domain-like receptors (NLRs; NLRC4, NLRP1b, or NLRP3) leads to loss of lysosomes via their fusion with the cell surface, or lysosome exocytosis. Active caspase-1 increased cellular membrane permeability and intracellular calcium levels, which facilitated lysosome exocytosis and release of host antimicrobial factors and microbial products. Lysosome exocytosis has been proposed to mediate secretion of IL-1β and IL-18; however, blocking lysosome exocytosis did not alter cytokine processing or release. These studies indicate two conserved secretion pathways are initiated by caspase-1, lysosome exocytosis, and a parallel pathway resulting in cytokine release, and both enhance the antimicrobial nature of pyroptosis.
Collapse
Affiliation(s)
- Tessa Bergsbaken
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
36
|
Type I interferon production induced by Streptococcus pyogenes-derived nucleic acids is required for host protection. PLoS Pathog 2011; 7:e1001345. [PMID: 21625574 PMCID: PMC3098218 DOI: 10.1371/journal.ppat.1001345] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 04/18/2011] [Indexed: 11/19/2022] Open
Abstract
Streptococcus pyogenes is a Gram-positive human pathogen that is recognized by yet unknown pattern recognition receptors (PRRs). Engagement of these receptor molecules during infection with S. pyogenes, a largely extracellular bacterium with limited capacity for intracellular survival, causes innate immune cells to produce inflammatory mediators such as TNF, but also type I interferon (IFN). Here we show that signaling elicited by type I IFNs is required for successful defense of mice against lethal subcutaneous cellulitis caused by S. pyogenes. Type I IFN signaling was accompanied with reduced neutrophil recruitment to the site of infection. Mechanistic analysis revealed that macrophages and conventional dendritic cells (cDCs) employ different signaling pathways leading to IFN-beta production. Macrophages required IRF3, STING, TBK1 and partially MyD88, whereas in cDCs the IFN-beta production was fully dependent on IRF5 and MyD88. Furthermore, IFN-beta production by macrophages was dependent on the endosomal delivery of streptococcal DNA, while in cDCs streptococcal RNA was identified as the IFN-beta inducer. Despite a role of MyD88 in both cell types, the known IFN-inducing TLRs were individually not required for generation of the IFN-beta response. These results demonstrate that the innate immune system employs several strategies to efficiently recognize S. pyogenes, a pathogenic bacterium that succeeded in avoiding recognition by the standard arsenal of TLRs. Streptococcus pyogenes is an important human pathogen that causes a broad range of diseases. The bacterium colonizes the throat and the skin where it can evoke usually mild illness such as strep throat or scarlet fever. Systemic infections with S. pyogenes are less frequent but can develop into life-threatening diseases such as necrotizing fasciitis and streptococcal toxic shock syndrome. The immune system launches a usually successful response that is initiated by a so far not understood recognition of this pathogen by the cells of the innate immune system. These cells produce upon infection a variety of cytokines that orchestrate a full blown protective response. Among these cytokines, type I interferons play a critical role as demonstrated by our study. We further show that IFN-beta, the key type I interferon, is produced only after macrophages and dendritic cells have taken up the pathogen and liberated the bacterial nucleic acids for recognition in the intracellular vesicles. Importantly, macrophages and dendritic cells recognize different nucleic acids and employ different signaling pathways to respond. Our data suggest that the innate immune system employs several strategies to efficiently recognize S. pyogenes, a pathogenic bacterium that succeeded in avoiding recognition by the standard recognition mechanisms.
Collapse
|
37
|
Abstract
Group A Streptococcus (GAS) can be internalized by epithelial cells, including keratinocytes from human skin or pharyngeal epithelium. Internalization of GAS by epithelial cells has been postulated both to play a role in host defense and to provide a sanctuary site for GAS survival. The cholesterol-binding cytolysin streptolysin O (SLO) appears to enhance virulence in part by inhibiting GAS internalization by human keratinocytes and by disrupting the lysosomal degradation of internalized GAS. We now report that low-level production of SLO by an inducible expression system reduced GAS internalization by keratinocytes. Induced SLO expression also prevented lysosomal colocalization with intracellular bacteria and acidification of GAS-containing vacuoles. Exogenous recombinant SLO mimicked the inhibitory effect of SLO secretion on GAS entry but not that on colocalization with the lysosomal marker LAMP-1, implying that disruption of lysosomal degradation requires intracellular secretion of SLO. The internalization of SLO-negative GAS was blocked by the depletion of host cell cholesterol and by the inhibition or knocking down of the expression of clathrin or dynamin. SLO also inhibited the cellular uptake of other cargos that are internalized by clathrin-mediated uptake or by macropinocytosis. We conclude that SLO interferes with the internalization of GAS through local perturbation of the keratinocyte cell membrane and disruption of a clathrin-dependent uptake pathway.
Collapse
|
38
|
Shelburne SA, Olsen RJ, Suber B, Sahasrabhojane P, Sumby P, Brennan RG, Musser JM. A combination of independent transcriptional regulators shapes bacterial virulence gene expression during infection. PLoS Pathog 2010; 6:e1000817. [PMID: 20333240 PMCID: PMC2841617 DOI: 10.1371/journal.ppat.1000817] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 02/09/2010] [Indexed: 11/18/2022] Open
Abstract
Transcriptional regulatory networks are fundamental to how microbes alter gene expression in response to environmental stimuli, thereby playing a critical role in bacterial pathogenesis. However, understanding how bacterial transcriptional regulatory networks function during host-pathogen interaction is limited. Recent studies in group A Streptococcus (GAS) suggested that the transcriptional regulator catabolite control protein A (CcpA) influences many of the same genes as the control of virulence (CovRS) two-component gene regulatory system. To provide new information about the CcpA and CovRS networks, we compared the CcpA and CovR transcriptomes in a serotype M1 GAS strain. The transcript levels of several of the same genes encoding virulence factors and proteins involved in basic metabolic processes were affected in both DeltaccpA and DeltacovR isogenic mutant strains. Recombinant CcpA and CovR bound with high-affinity to the promoter regions of several co-regulated genes, including those encoding proteins involved in carbohydrate and amino acid metabolism. Compared to the wild-type parental strain, DeltaccpA and DeltacovRDeltaccpA isogenic mutant strains were significantly less virulent in a mouse myositis model. Inactivation of CcpA and CovR alone and in combination led to significant alterations in the transcript levels of several key GAS virulence factor encoding genes during infection. Importantly, the transcript level alterations in the DeltaccpA and DeltacovRDeltaccpA isogenic mutant strains observed during infection were distinct from those occurring during growth in laboratory medium. These data provide new knowledge regarding the molecular mechanisms by which pathogenic bacteria respond to environmental signals to regulate virulence factor production and basic metabolic processes during infection.
Collapse
Affiliation(s)
- Samuel A. Shelburne
- Department of Infectious Diseases, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Randall J. Olsen
- Center for Molecular and Translational Human Infectious Diseases Research, The Methodist Hospital Research Institute, and Department of Pathology, The Methodist Hospital, Houston, Texas, United States of America
| | - Bryce Suber
- Department of Infectious Diseases, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Pranoti Sahasrabhojane
- Department of Infectious Diseases, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Paul Sumby
- Center for Molecular and Translational Human Infectious Diseases Research, The Methodist Hospital Research Institute, and Department of Pathology, The Methodist Hospital, Houston, Texas, United States of America
| | - Richard G. Brennan
- Department of Biochemistry and Molecular Biology, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - James M. Musser
- Center for Molecular and Translational Human Infectious Diseases Research, The Methodist Hospital Research Institute, and Department of Pathology, The Methodist Hospital, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Cortés G, Wessels MR. Inhibition of dendritic cell maturation by group A Streptococcus. J Infect Dis 2009; 200:1152-61. [PMID: 19712038 DOI: 10.1086/605696] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Exposure to group A Streptococcus (GAS) has been shown to induce maturation of dendritic cells (DC). METHODS To identify bacterial determinants that modulate DC activation in response to GAS infection, we analyzed the induction of maturation in human monocyte-derived DC following exposure to GAS clinical isolates. RESULTS Unexpectedly, only 6 of 24 GAS strains tested induced surface expression of major histocompatibility complex class II and costimulatory molecules CD80 and CD83 to levels consistent with DC maturation. Rather, the majority of the strains did not promote DC maturation, and many triggered DC apoptosis. GAS strains that failed to induce DC maturation were those that produced abundant hyaluronic acid (HA) capsular polysaccharide and/or large amounts of the cytotoxin streptolysin O (SLO). By use of isogenic mutants deficient in HA and/or SLO, we determined that GAS inhibits DC maturation through 2 distinct mechanisms: (1) inhibition of bacterial binding and/or phagocytosis by the HA capsule and (2) SLO-mediated induction of DC apoptosis by intracellular GAS. CONCLUSIONS These results demonstrate that GAS virulence factors modulate maturation and survival of human DC, effects that are likely to have a critical impact on activation of innate and adaptive immune responses to this important human pathogen.
Collapse
Affiliation(s)
- Guadalupe Cortés
- Division of Infectious Diseases, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
40
|
Nobbs AH, Lamont RJ, Jenkinson HF. Streptococcus adherence and colonization. Microbiol Mol Biol Rev 2009; 73:407-50, Table of Contents. [PMID: 19721085 PMCID: PMC2738137 DOI: 10.1128/mmbr.00014-09] [Citation(s) in RCA: 437] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Streptococci readily colonize mucosal tissues in the nasopharynx; the respiratory, gastrointestinal, and genitourinary tracts; and the skin. Each ecological niche presents a series of challenges to successful colonization with which streptococci have to contend. Some species exist in equilibrium with their host, neither stimulating nor submitting to immune defenses mounted against them. Most are either opportunistic or true pathogens responsible for diseases such as pharyngitis, tooth decay, necrotizing fasciitis, infective endocarditis, and meningitis. Part of the success of streptococci as colonizers is attributable to the spectrum of proteins expressed on their surfaces. Adhesins enable interactions with salivary, serum, and extracellular matrix components; host cells; and other microbes. This is the essential first step to colonization, the development of complex communities, and possible invasion of host tissues. The majority of streptococcal adhesins are anchored to the cell wall via a C-terminal LPxTz motif. Other proteins may be surface anchored through N-terminal lipid modifications, while the mechanism of cell wall associations for others remains unclear. Collectively, these surface-bound proteins provide Streptococcus species with a "coat of many colors," enabling multiple intimate contacts and interplays between the bacterial cell and the host. In vitro and in vivo studies have demonstrated direct roles for many streptococcal adhesins as colonization or virulence factors, making them attractive targets for therapeutic and preventive strategies against streptococcal infections. There is, therefore, much focus on applying increasingly advanced molecular techniques to determine the precise structures and functions of these proteins, and their regulatory pathways, so that more targeted approaches can be developed.
Collapse
Affiliation(s)
- Angela H Nobbs
- Oral Microbiology Unit, Department of Oral and Dental Science, University of Bristol, Bristol BS1 2LY, United Kingdom
| | | | | |
Collapse
|
41
|
Russell HH, Zhou L, Sriskandan S. Rapid screen for epithelial internalization of Tn917-mutagenized Streptococcus pyogenes. J Microbiol Methods 2009; 78:34-9. [PMID: 19371765 DOI: 10.1016/j.mimet.2009.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 04/03/2009] [Accepted: 04/06/2009] [Indexed: 10/20/2022]
Abstract
Group A streptococci (GAS) cause a number of human diseases ranging from pharyngitis to necrotizing fasciitis. GAS are hypothesized to escape killing by either the immune system or beta lactam antibiotics by internalization into epithelial cells. A Tn917 library of transposon mutants was screened for capacity to invade and survive in human epithelial cells using a novel blood agar overlay method. Although the screen revealed that a majority of Tn917 insertions occurred within a 10 kb region of the genome, GAS genes identified as essential for internalization into epithelial cells included ABC transporters, and DNA maintenance proteins, and citrate metabolism enzymes, underlining the importance of adaptation to the intracellular environment.
Collapse
Affiliation(s)
- Hugh H Russell
- Department of Infectious Diseases & Immunity, Hammersmith Campus, Imperial College London, UK
| | | | | |
Collapse
|
42
|
Polysaccharide capsule and suilysin contribute to extracellular survival of Streptococcus suis co-cultivated with primary porcine phagocytes. Vet Microbiol 2008; 132:211-9. [DOI: 10.1016/j.vetmic.2008.05.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 04/29/2008] [Accepted: 05/05/2008] [Indexed: 11/19/2022]
|
43
|
Radtke AL, O'Riordan MXD. Homeostatic maintenance of pathogen-containing vacuoles requires TBK1-dependent regulation of aquaporin-1. Cell Microbiol 2008; 10:2197-207. [PMID: 18665841 DOI: 10.1111/j.1462-5822.2008.01199.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Membranes are an integral component of many cellular functions and serve as a barrier to keep pathogenic bacteria from entering the nutrient-rich host cytosol. TANK-binding-kinase-1 (TBK1), a kinase of the IkappaB kinase family, is required for maintaining integrity of pathogen-containing vacuoles (PCV) upon bacterial invasion of host cells. Here we investigate how vacuolar integrity is maintained during bacterial infection, even in the presence of bacterial membrane damaging agents. We found that Aquaporin-1 (AQP1), a water channel that regulates swelling of secretory vesicles, associated with PCV. AQP1 levels were elevated in TBK1-deficient cells, and overexpression of AQP1 in wild-type cells led to PCV destabilization, similar to that observed in tbk1(-/-) cells. Inhibition of physiological levels of AQP1 in multiple cell types also led to increased instability of PCV, demonstrating a need for tightly regulated AQP1 function to maintain vacuole homeostasis during bacterial infection. AQP1-dependent modulation of PCV was triggered by bacterially induced membrane damage and ion flux. These results highlight the contribution of water channels to promoting PCV membrane integrity, and reveal an unexpected role for TBK1 and AQP1 in restricting bacterial pathogens to the vacuolar compartment.
Collapse
Affiliation(s)
- Andrea L Radtke
- Department of Microbiology and Immunology, University of Michigan Medical School, 1150 W. Medical Center Drive, 5641 Medical Sciences II, Ann Arbor, MI 48109-5620, USA
| | | |
Collapse
|
44
|
Ryan PA, Kirk BW, Euler CW, Schuch R, Fischetti VA. Novel algorithms reveal streptococcal transcriptomes and clues about undefined genes. PLoS Comput Biol 2008; 3:e132. [PMID: 17616984 PMCID: PMC1913099 DOI: 10.1371/journal.pcbi.0030132] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Accepted: 05/22/2007] [Indexed: 01/22/2023] Open
Abstract
Bacteria-host interactions are dynamic processes, and understanding transcriptional responses that directly or indirectly regulate the expression of genes involved in initial infection stages would illuminate the molecular events that result in host colonization. We used oligonucleotide microarrays to monitor (in vitro) differential gene expression in group A streptococci during pharyngeal cell adherence, the first overt infection stage. We present neighbor clustering, a new computational method for further analyzing bacterial microarray data that combines two informative characteristics of bacterial genes that share common function or regulation: (1) similar gene expression profiles (i.e., co-expression); and (2) physical proximity of genes on the chromosome. This method identifies statistically significant clusters of co-expressed gene neighbors that potentially share common function or regulation by coupling statistically analyzed gene expression profiles with the chromosomal position of genes. We applied this method to our own data and to those of others, and we show that it identified a greater number of differentially expressed genes, facilitating the reconstruction of more multimeric proteins and complete metabolic pathways than would have been possible without its application. We assessed the biological significance of two identified genes by assaying deletion mutants for adherence in vitro and show that neighbor clustering indeed provides biologically relevant data. Neighbor clustering provides a more comprehensive view of the molecular responses of streptococci during pharyngeal cell adherence.
Collapse
Affiliation(s)
- Patricia A Ryan
- Department of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, New York, USA.
| | | | | | | | | |
Collapse
|
45
|
Koo IC, Wang C, Raghavan S, Morisaki JH, Cox JS, Brown EJ. ESX-1-dependent cytolysis in lysosome secretion and inflammasome activation during mycobacterial infection. Cell Microbiol 2008; 10:1866-78. [PMID: 18503637 DOI: 10.1111/j.1462-5822.2008.01177.x] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Exocytosis of lysosomes from macrophages has been described as a response to microbial cytotoxins and haemolysins, as well as for releasing pro-inflammatory cytokines interleukin (IL)-1beta and IL-18 during inflammasome activation. The mycobacterial ESX-1 secretion system, encoded in part by the Region of Difference-1, is a virulence factor necessary for phagosome escape and host cell lysis by a contact-dependent haemolysin in Mycobacterium marinum. Here we show that ESX-1 from M. marinum and M. tuberculosis is required for Ca(2+)-dependent induction of lysosome secretion from macrophages. Mycobacteria-induced lysosome secretion was concurrent to release of IL-1beta and IL-18, dependent on phagocytosis of bacteria containing ESX-1. Synthesis but not release of IL-1beta and IL-18 occurred in response to dead bacilli and bacteria lacking ESX-1, indicating that only cytokine release was regulated by ESX-1. Release of these cytokines and exocytosis of lysosomes were independent of intracellular mycobacterial growth, yet correlated with mycobacteria-encoded haemolytic activity, demonstrating a parallel pathway for the two responses. We further identified inflammasome components caspase-1, ASC and NALP3, but not Ipaf, required for release of IL-1beta and IL-18. Collectively, these results reveal a role for ESX-1 in triggering secretion of lysosomes, as well as release of IL-1beta and IL-18 during mycobacteria infection.
Collapse
Affiliation(s)
- Ingrid C Koo
- Program in Microbial Pathogenesis and Host Defense, University of California, San Fancisco, CA, USA
| | | | | | | | | | | |
Collapse
|
46
|
Barbuddhe S, Chakraborty T. Biotechnological applications of Listeria's sophisticated infection strategies. Microb Biotechnol 2008; 1:361-72. [PMID: 21261856 PMCID: PMC3815243 DOI: 10.1111/j.1751-7915.2008.00037.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Listeria monocytogenes is a Gram‐positive bacterium that is able to survive both in the environment and to invade and multiply within eukaryotic cells. Currently L. monocytogenes represents one of the most well‐studied and characterized microorganisms in bacterial pathogenesis. A hallmark of L. monocytogenes virulence is its ability to breach bodily barriers such as the intestinal epithelium, the blood–brain barrier as well as the placental barrier to cause severe systemic disease. Curiously, this theme is repeated at the level of the interaction between the individual cell and the bacterium where its virulence factors contribute to the ability of the bacteria to breach cellular barriers. L. monocytogenes is a model to study metabolic requirements of bacteria growing in an intracellular environment, modulation of signalling pathways in the infected cell and interactions with cellular defences involving innate and adaptive immunity. Technical advances such as the creation of LISTERIA‐susceptible mouse strains, had added interest in the study of the natural pathogenesis of the disease via oral infection. The use of attenuated strains of L. monocytogenes as vaccines has gained considerable interest because they can be used to express heterologous antigens as well as to somatically deliver recombinant DNA to eukaryotic cells. A novel vaccine concept, the use of non‐viable but metabolically active bacteria to induced immunoprotective responses, has been developed with L. monocytogenes. In this mini‐review, we review the strategies used by L. monocytogenes to subvert the cellular functions at different stages of the infection cycle in the host and examine how these properties are being exploited in biotechnological and clinical applications.
Collapse
Affiliation(s)
- Sukhadeo Barbuddhe
- Institute for Medical Microbiology, Justus-Liebig University, Frankfurter strasse 107, D-35392 Giessen, Germany
| | | |
Collapse
|
47
|
Gratz N, Siller M, Schaljo B, Pirzada ZA, Gattermeier I, Vojtek I, Kirschning CJ, Wagner H, Akira S, Charpentier E, Kovarik P. Group A streptococcus activates type I interferon production and MyD88-dependent signaling without involvement of TLR2, TLR4, and TLR9. J Biol Chem 2008; 283:19879-87. [PMID: 18480050 PMCID: PMC2459277 DOI: 10.1074/jbc.m802848200] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bacterial pathogens are recognized by the innate immune system through pattern recognition receptors, such as Toll-like receptors (TLRs). Engagement of TLRs triggers signaling cascades that launch innate immune responses. Activation of MAPKs and NF-kappaB, elements of the major signaling pathways induced by TLRs, depends in most cases on the adaptor molecule MyD88. In addition, Gram-negative or intracellular bacteria elicit MyD88-independent signaling that results in production of type I interferon (IFN). Here we show that in mouse macrophages, the activation of MyD88-dependent signaling by the extracellular Gram-positive human pathogen group A streptococcus (GAS; Streptococcus pyogenes) does not require TLR2, a receptor implicated in sensing of Gram-positive bacteria, or TLR4 and TLR9. Redundant engagement of either of these TLR molecules was excluded by using TLR2/4/9 triple-deficient macrophages. We further demonstrate that infection of macrophages by GAS causes IRF3 (interferon-regulatory factor 3)-dependent, MyD88-independent production of IFN. Surprisingly, IFN is induced also by GAS lacking slo and sagA, the genes encoding cytolysins that were shown to be required for IFN production in response to other Gram-positive bacteria. Our data indicate that (i) GAS is recognized by a MyD88-dependent receptor other than any of those typically used by bacteria, and (ii) GAS as well as GAS mutants lacking cytolysin genes induce type I IFN production by similar mechanisms as bacteria requiring cytoplasmic escape and the function of cytolysins.
Collapse
Affiliation(s)
- Nina Gratz
- Max F Perutz Laboratories, Department of Microbiology and Immunobiology, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Angus AA, Lee AA, Augustin DK, Lee EJ, Evans DJ, Fleiszig SMJ. Pseudomonas aeruginosa induces membrane blebs in epithelial cells, which are utilized as a niche for intracellular replication and motility. Infect Immun 2008; 76:1992-2001. [PMID: 18316391 PMCID: PMC2346716 DOI: 10.1128/iai.01221-07] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 10/10/2007] [Accepted: 02/14/2008] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is known to invade epithelial cells during infection and in vitro. However, little is known of bacterial or epithelial factors modulating P. aeruginosa intracellular survival or replication after invasion, except that it requires a complete lipopolysaccharide core. In this study, real-time video microscopy revealed that invasive P. aeruginosa isolates induced the formation of membrane blebs in multiple epithelial cell types and that these were then exploited for intracellular replication and rapid real-time motility. Further studies revealed that the type three secretion system (T3SS) of P. aeruginosa was required for blebbing. Mutants lacking either the entire T3SS or specific T3SS components were instead localized to intracellular perinuclear vacuoles. Most T3SS mutants that trafficked to perinuclear vacuoles gradually lost intracellular viability, and vacuoles containing those bacteria were labeled by the late endosomal marker lysosome-associated marker protein 3 (LAMP-3). Interestingly, mutants deficient only in the T3SS translocon structure survived and replicated within the vacuoles that did not label with LAMP-3. Taken together, these data suggest two novel roles of the P. aeruginosa T3SS in enabling bacterial intracellular survival: translocon-dependent formation of membrane blebs, which form a host cell niche for bacterial growth and motility, and effector-dependent bacterial survival and replication within intracellular perinuclear vacuoles.
Collapse
Affiliation(s)
- Annette A Angus
- School of Optometry, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | |
Collapse
|
49
|
Role for lysosomal enzyme beta-hexosaminidase in the control of mycobacteria infection. Proc Natl Acad Sci U S A 2008; 105:710-5. [PMID: 18180457 DOI: 10.1073/pnas.0708110105] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The pathogenic mycobacteria that cause tuberculosis (TB) and TB-like diseases in humans and animals elude sterilizing immunity by residing within an intracellular niche in host macrophages, where they are protected from microbicidal attack. Recent studies have emphasized microbial mechanisms for evasion of host defense; less is known about mycobactericidal mechanisms that remain intact during initial infection. To better understand macrophage mechanisms for restricting mycobacteria growth, we examined Mycobacterium marinum infection of Drosophila S2 cells. Among approximately 1,000 host genes examined by RNAi depletion, the lysosomal enzyme beta-hexosaminidase was identified as an important factor in the control of mycobacterial infection. The importance of beta-hexosaminidase for restricting mycobacterial growth during mammalian infections was confirmed in macrophages from beta-hexosaminidase knockout mice. Beta-hexosaminidase was characterized as a peptidoglycan hydrolase that surprisingly exerts its mycobactericidal effect at the macrophage plasma membrane during mycobacteria-induced secretion of lysosomes. Thus, secretion of lysosomal enzymes is a mycobactericidal mechanism that may have a more general role in host defense.
Collapse
|
50
|
Abstract
Group A Streptococcus (GAS) is a Gram-positive bacterium associated with a variety of mucosal and invasive human infections. GAS systemic disease reflects the diverse abilities of this pathogen to avoid eradication by phagocytic defenses of the innate immune system. Here we review how GAS can avoid phagocyte engagement, inhibit complement and antibody functions required for opsonization, impair phagocytotic uptake mechanisms, promote phagocyte lysis or apoptosis, and resist specific effectors of phagocyte killing such as antimicrobial peptides and reactive oxygen species. Understanding the molecular basis of GAS phagocyte resistance may reveal novel therapeutic targets for treatment and prevention of invasive human infections.
Collapse
Affiliation(s)
- Laura A Kwinn
- Division of Pediatric Pharmacology & Drug Discovery, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA.
| | | |
Collapse
|