1
|
Zhu Z, Hu Z, Li S, Fang R, Ono HK, Hu DL. Molecular Characteristics and Pathogenicity of Staphylococcus aureus Exotoxins. Int J Mol Sci 2023; 25:395. [PMID: 38203566 PMCID: PMC10778951 DOI: 10.3390/ijms25010395] [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: 11/24/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Staphylococcus aureus stands as one of the most pervasive pathogens given its morbidity and mortality worldwide due to its roles as an infectious agent that causes a wide variety of diseases ranging from moderately severe skin infections to fatal pneumonia and sepsis. S. aureus produces a variety of exotoxins that serve as important virulence factors in S. aureus-related infectious diseases and food poisoning in both humans and animals. For example, staphylococcal enterotoxins (SEs) produced by S. aureus induce staphylococcal foodborne poisoning; toxic shock syndrome toxin-1 (TSST-1), as a typical superantigen, induces toxic shock syndrome; hemolysins induce cell damage in erythrocytes and leukocytes; and exfoliative toxin induces staphylococcal skin scalded syndrome. Recently, Panton-Valentine leucocidin, a cytotoxin produced by community-associated methicillin-resistant S. aureus (CA-MRSA), has been reported, and new types of SEs and staphylococcal enterotoxin-like toxins (SEls) were discovered and reported successively. This review addresses the progress of and novel insights into the molecular structure, biological activities, and pathogenicity of both the classic and the newly identified exotoxins produced by S. aureus.
Collapse
Affiliation(s)
- Zhihao Zhu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zuo Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Shaowen Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Rendong Fang
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China;
| | - Hisaya K. Ono
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Dong-Liang Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| |
Collapse
|
2
|
Abstract
Neutrophils or polymorphonuclear neutrophils (PMNs) are an important component of innate host defense. These phagocytic leukocytes are recruited to infected tissues and kill invading microbes. There are several general characteristics of neutrophils that make them highly effective as antimicrobial cells. First, there is tremendous daily production and turnover of granulocytes in healthy adults-typically 1011 per day. The vast majority (~95%) of these cells are neutrophils. In addition, neutrophils are mobilized rapidly in response to chemotactic factors and are among the first leukocytes recruited to infected tissues. Most notably, neutrophils contain and/or produce an abundance of antimicrobial molecules. Many of these antimicrobial molecules are toxic to host cells and can destroy host tissues. Thus, neutrophil activation and turnover are highly regulated processes. To that end, aged neutrophils undergo apoptosis constitutively, a process that contains antimicrobial function and proinflammatory capacity. Importantly, apoptosis facilitates nonphlogistic turnover of neutrophils and removal by macrophages. This homeostatic process is altered by interaction with microbes and their products, as well as host proinflammatory molecules. Microbial pathogens can delay neutrophil apoptosis, accelerate apoptosis following phagocytosis, or cause neutrophil cytolysis. Here, we review these processes and provide perspective on recent studies that have potential to impact this paradigm.
Collapse
Affiliation(s)
- Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Mark T Quinn
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
3
|
Cheung GYC, Bae JS, Otto M. Pathogenicity and virulence of Staphylococcus aureus. Virulence 2021; 12:547-569. [PMID: 33522395 PMCID: PMC7872022 DOI: 10.1080/21505594.2021.1878688] [Citation(s) in RCA: 502] [Impact Index Per Article: 167.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is one of the most frequent worldwide causes of morbidity and mortality due to an infectious agent. This pathogen can cause a wide variety of diseases, ranging from moderately severe skin infections to fatal pneumonia and sepsis. Treatment of S. aureus infections is complicated by antibiotic resistance and a working vaccine is not available. There has been ongoing and increasing interest in the extraordinarily high number of toxins and other virulence determinants that S. aureus produces and how they impact disease. In this review, we will give an overview of how S. aureus initiates and maintains infection and discuss the main determinants involved. A more in-depth understanding of the function and contribution of S. aureus virulence determinants to S. aureus infection will enable us to develop anti-virulence strategies to counteract the lack of an anti-S. aureus vaccine and the ever-increasing shortage of working antibiotics against this important pathogen.
Collapse
Affiliation(s)
- Gordon Y. C. Cheung
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, Maryland, USA
| | - Justin S. Bae
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
4
|
Wurtz M, Ruhland E, Liu X, Namer IJ, Mazzoleni V, Lipsker D, Keller D, Prévost G, Gaucher D. Panton-Valentine Leucocidin of Staphylococcus aureus Induces Oxidative Stress and Neurotransmitter Imbalance in a Retinal Explant Model. Invest Ophthalmol Vis Sci 2021; 62:4. [PMID: 33393970 PMCID: PMC7794257 DOI: 10.1167/iovs.62.1.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Endophthalmitis models have reported the virulent role of Panton-Valentine leucocidin (PVL) secreted by Staphylococcus aureus on the retina. PVL targets retinal ganglion cells (RGCs), expressing PVL membrane receptor C5aR. Interactions between PVL and retinal cells lead to glial activation, retinal inflammation, and apoptosis. In this study, we explored oxidative stress and retinal neurotransmitters in a rabbit retinal explant model incubated with PVL. Methods Reactive oxygen species (ROS) production in RGCs has been assessed with fluorescent probes and immunohistochemistry. Nuclear magnetic resonance (NMR) spectroscopy quantified retinal concentrations of antioxidant molecules and neurotransmitters, and concentrations of neurotransmitters released in the culture medium. Quantifying the expression of some pro-inflammatory and anti-inflammatory factors was performed using RT-qPCR. Results PVL induced a mitochondrial ROS production in RGCs after four hours’ incubation with the toxin. Enzymatic sources of ROS, involving nicotinamide adenine dinucleotide phosphate–oxidase and xanthine oxidase, were also activated after four hours in PVL-treated retinal explants. Retinal antioxidants defenses, that is, glutathione, ascorbate and taurine, decreased after two hours’ incubation with PVL. Glutamate retinal concentrations and glutamate release in the culture medium remained unaltered in PVL-treated retinas. GABA, glycine, and acetylcholine (Ach) retinal concentrations decreased after PVL treatment. Glycine release in the culture medium decreased, whereas Ach release increased after PVL treatment. Expression of proinflammatory and anti-inflammatory cytokines remained unchanged in PVL-treated explants. Conclusions PVL activates oxidative pathways and alters neurotransmitter retinal concentrations and release, supporting the hypothesis that PVL could induce a neurogenic inflammation in the retina.
Collapse
Affiliation(s)
- Mathieu Wurtz
- University of Strasbourg, Hôpitaux Universitaires de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, UR7290 Virulence Bactérienne Précoce, Institute of Bacteriology, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Department of Ophthalmology, Nouvel Hôpital Civil, Strasbourg, France
| | - Elisa Ruhland
- MNMS Platform, Department of Biophysics and Nuclear Medicine, Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Strasbourg, France
| | - XuanLi Liu
- University of Strasbourg, Hôpitaux Universitaires de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, UR7290 Virulence Bactérienne Précoce, Institute of Bacteriology, Strasbourg, France
| | - Izzie-Jacques Namer
- MNMS Platform, Department of Biophysics and Nuclear Medicine, Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Strasbourg, France
| | - Viola Mazzoleni
- University of Strasbourg, Hôpitaux Universitaires de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, UR7290 Virulence Bactérienne Précoce, Institute of Bacteriology, Strasbourg, France
| | - Dan Lipsker
- Hôpitaux Universitaires de Strasbourg, Department of Dermatology, Nouvel Hôpital Civil, Strasbourg, France
| | - Daniel Keller
- University of Strasbourg, Hôpitaux Universitaires de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, UR7290 Virulence Bactérienne Précoce, Institute of Bacteriology, Strasbourg, France
| | - Gilles Prévost
- University of Strasbourg, Hôpitaux Universitaires de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, UR7290 Virulence Bactérienne Précoce, Institute of Bacteriology, Strasbourg, France
| | - David Gaucher
- University of Strasbourg, Hôpitaux Universitaires de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, UR7290 Virulence Bactérienne Précoce, Institute of Bacteriology, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Department of Ophthalmology, Nouvel Hôpital Civil, Strasbourg, France
| |
Collapse
|
5
|
Mazzoleni V, Zimmermann K, Smirnova A, Tarassov I, Prévost G. Staphylococcus aureus Panton-Valentine Leukocidin triggers an alternative NETosis process targeting mitochondria. FASEB J 2020; 35:e21167. [PMID: 33241563 DOI: 10.1096/fj.201902981r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 09/18/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Panton-Valentine Leukocidin (PVL) is a bicomponent leukotoxin produced by 3%-10% of clinical Staphylococcus aureus (SA) strains involved in the severity of hospital and community-acquired infections. Although PVL was long known as a pore-forming toxin, recent studies have challenged the formation of a pore at the plasma membrane, while its endocytosis and the exact mode of action remain to be defined. In vitro immunolabeling of human neutrophils shows that Neutrophil Extracellular Traps (NETosis) is triggered by the action of purified PVL, but not by Gamma hemolysin CB (HlgCB), a structurally similar SA leukotoxin. PVL causes the ejection of chromatin fibers (NETs) decorated with antibacterial peptides independently of the NADPH oxidase oxidative burst. Leukotoxin partially colocalizes with mitochondria and enhances the production of reactive oxygen species from these organelles, while showing an increased autophagy, which results unnecessary for NETs ejection. PVL NETosis is elicited through Ca2+ -activated SK channels and Myeloperoxidase activity but is abolished by Allopurinol pretreatment of neutrophils. Moreover, massive citrullination of the histone H3 is performed by peptidyl arginine deiminases. Inhibition of this latter enzymes fails to abolish NET extrusion. Unexpectedly, PVL NETosis does not seem to involve Src kinases, which is the main kinase family activated downstream the binding of PVL F subunit to CD45 receptor, while the specific kinase pathway differs from the NADPH oxidase-dependent NETosis. PVL alone causes a different and specific form of NETosis that may rather represent a bacterial strategy conceived to disarm and disrupt the immune response, eventually allowing SA to spread.
Collapse
Affiliation(s)
- Viola Mazzoleni
- University of Strasbourg, CHRU Strasbourg, ITI InnoVec, Fédération de Médecine Translationnelle de Strasbourg, UR7290, Institut de Bactériologie, Strasbourg, France
| | - Kiran Zimmermann
- University of Strasbourg, CHRU Strasbourg, ITI InnoVec, Fédération de Médecine Translationnelle de Strasbourg, UR7290, Institut de Bactériologie, Strasbourg, France
| | - Anna Smirnova
- UMR 7156 GMGM Strasbourg University/CNRS, Strasbourg, France
| | - Ivan Tarassov
- UMR 7156 GMGM Strasbourg University/CNRS, Strasbourg, France
| | - Gilles Prévost
- University of Strasbourg, CHRU Strasbourg, ITI InnoVec, Fédération de Médecine Translationnelle de Strasbourg, UR7290, Institut de Bactériologie, Strasbourg, France
| |
Collapse
|
6
|
Forbes JD. Clinically Important Toxins in Bacterial Infection: Utility of Laboratory Detection. CLINICAL MICROBIOLOGY NEWSLETTER 2020; 42:163-170. [PMID: 33046946 PMCID: PMC7541054 DOI: 10.1016/j.clinmicnews.2020.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The elaboration of proteins that damage host cells is fundamental to the pathogenesis of many bacterial pathogens. The clinical significance of many bacterial toxins is well recognized, and routine detection is necessary to confirm definitive diagnosis for some types of infectious diseases. Determining the clinical significance of a toxin involves many factors, including the toxin's prevalence, virulence, and role in disease pathogenesis. While essential from a diagnostic perspective, toxin detection has the potential to be important for patient management decision making, as well as infection prevention and control measures. This review focuses on the history, epidemiology, pathogenesis, clinical presentation, and management of infections associated with well-defined, clinically important toxins (such as Shiga toxin-producing Escherichia coli), as well as those that are less well defined (such as Staphylococcus aureus' Panton-Valentine leukocidin) where detection may yield clinically important information.
Collapse
Affiliation(s)
- Jessica D Forbes
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
7
|
Transcriptional Profiling Suggests T Cells Cluster around Neurons Injected with Toxoplasma gondii Proteins. mSphere 2020; 5:5/5/e00538-20. [PMID: 32878927 PMCID: PMC7471001 DOI: 10.1128/msphere.00538-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Like other persistent intracellular pathogens, Toxoplasma gondii, a protozoan parasite, has evolved to evade the immune system and establish a chronic infection in specific cells and organs, including neurons in the CNS. Understanding T. gondii’s persistence in neurons holds the potential to identify novel, curative drug targets. The work presented here offers new insights into the neuron-T. gondii interaction in vivo. By transcriptionally profiling neurons manipulated by T. gondii, we unexpectedly revealed that immune cells, and specifically CD8+ T cells, appear to cluster around these neurons, suggesting that CD8+ T cells specifically recognize parasite-manipulated neurons. Such a possibility supports evidence from other labs that questions the long-standing dogma that neurons are often persistently infected because they are not directly recognized by immune cells such as CD8+ T cells. Collectively, these data suggest we reconsider the broader role of neurons in the context of infection and neuroinflammation. Toxoplasma gondii’s tropism for and persistence in the central nervous system (CNS) underlies the symptomatic disease that T. gondii causes in humans. Our recent work has shown that neurons are the primary CNS cell with which Toxoplasma interacts and which it infects in vivo. This predilection for neurons suggests that T. gondii’s persistence in the CNS depends specifically upon parasite manipulation of the host neurons. Yet, most work on T. gondii-host cell interactions has been done in vitro and in nonneuronal cells. We address this gap by utilizing our T. gondii-Cre system that allows permanent marking and tracking of neurons injected with parasite effector proteins in vivo. Using laser capture microdissection (LCM) and RNA sequencing using RNA-seq, we isolated and transcriptionally profiled T. gondii-injected neurons (TINs), Bystander neurons (nearby non-T. gondii-injected neurons), and neurons from uninfected mice (controls). These profiles show that TIN transcriptomes significantly differ from the transcriptomes of Bystander and control neurons and that much of this difference is driven by increased levels of transcripts from immune cells, especially CD8+ T cells and monocytes. These data suggest that when we used LCM to isolate neurons from infected mice, we also picked up fragments of CD8+ T cells and monocytes clustering in extreme proximity around TINs and, to a lesser extent, Bystander neurons. In addition, we found that T. gondii transcripts were primarily found in the TIN transcriptome, not in the Bystander transcriptome. Collectively, these data suggest that, contrary to common perception, neurons that directly interact with or harbor parasites can be recognized by CD8+ T cells. IMPORTANCE Like other persistent intracellular pathogens, Toxoplasma gondii, a protozoan parasite, has evolved to evade the immune system and establish a chronic infection in specific cells and organs, including neurons in the CNS. Understanding T. gondii’s persistence in neurons holds the potential to identify novel, curative drug targets. The work presented here offers new insights into the neuron-T. gondii interaction in vivo. By transcriptionally profiling neurons manipulated by T. gondii, we unexpectedly revealed that immune cells, and specifically CD8+ T cells, appear to cluster around these neurons, suggesting that CD8+ T cells specifically recognize parasite-manipulated neurons. Such a possibility supports evidence from other labs that questions the long-standing dogma that neurons are often persistently infected because they are not directly recognized by immune cells such as CD8+ T cells. Collectively, these data suggest we reconsider the broader role of neurons in the context of infection and neuroinflammation.
Collapse
|
8
|
Bennett MR, Thomsen IP. Epidemiological and Clinical Evidence for the Role of Toxins in S. aureus Human Disease. Toxins (Basel) 2020; 12:toxins12060408. [PMID: 32575633 PMCID: PMC7354447 DOI: 10.3390/toxins12060408] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus asymptomatically colonizes approximately 30–50% of the population and is a leading cause of bacteremia, bone/joint infections, and skin infections in the US. S. aureus has become a major public health threat due to antibiotic resistance and an increasing number of failed vaccine attempts. To develop new anti-staphylococcal preventive therapies, it will take a more thorough understanding of the current role S. aureus virulence factors play in contributing to human disease. This review focuses on the clinical association of individual toxins with S. aureus infection as well as attempted treatment options. Further understanding of these associations will increase understanding of toxins and their importance to S. aureus pathogenesis.
Collapse
Affiliation(s)
- Monique R. Bennett
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
- Vanderbilt Vaccine Research Program, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Isaac P. Thomsen
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
- Vanderbilt Vaccine Research Program, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
9
|
Sward EW, Fones EM, Spaan RR, Pallister KB, Haller BL, Guerra FE, Zurek OW, Nygaard TK, Voyich JM. Staphylococcus aureus SaeR/S-Regulated Factors Decrease Monocyte-Derived Tumor Necrosis Factor-α to Reduce Neutrophil Bactericidal Activity. J Infect Dis 2019; 217:943-952. [PMID: 29272502 DOI: 10.1093/infdis/jix652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/17/2017] [Indexed: 12/11/2022] Open
Abstract
Background The ability of Staphylococcus aureus to evade killing by human neutrophils significantly contributes to disease progression. In this study, we characterize an influential role for the S. aureus SaeR/S 2-component gene regulatory system in suppressing monocyte production of tumor necrosis factor alpha (TNF-α) to subsequently influence human neutrophil priming. Methods Using flow cytometry and TNF-α specific enzyme-linked immunosorbent assays we identify the primary cellular source of TNF-α in human blood and in purified peripheral blood mononuclear cells (PBMCs) during interaction with USA300 and an isogenic saeR/S deletion mutant (USA300∆saeR/S). Assays with conditioned media from USA300 and USA300∆saeR/S exposed PBMCs were used to investigate priming on neutrophil bactericidal activity. Results TNF-α production from monocytes was significantly reduced following challenge with USA300 compared to USA300∆saeR/S. We observed that priming of neutrophils using conditioned medium from peripheral blood mononuclear cells stimulated with USA300∆saeR/S significantly increased neutrophil bactericidal activity against USA300 relative to unprimed neutrophils and neutrophils primed with USA300 conditioned medium. The increased neutrophil bactericidal activity was associated with enhanced reactive oxygen species production that was significantly influenced by elevated TNF-α concentrations. Conclusions Our findings identify an immune evasion strategy used by S. aureus to impede neutrophil priming and subsequent bactericidal activity.
Collapse
Affiliation(s)
- Eli W Sward
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Elizabeth M Fones
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Russel R Spaan
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Brandon L Haller
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Fermin E Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Oliwia W Zurek
- Infectious Disease Department, Genentech Inc, South San Francisco, California
| | - Tyler K Nygaard
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman
| |
Collapse
|
10
|
The Role of Streptococcal and Staphylococcal Exotoxins and Proteases in Human Necrotizing Soft Tissue Infections. Toxins (Basel) 2019; 11:toxins11060332. [PMID: 31212697 PMCID: PMC6628391 DOI: 10.3390/toxins11060332] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 12/31/2022] Open
Abstract
Necrotizing soft tissue infections (NSTIs) are critical clinical conditions characterized by extensive necrosis of any layer of the soft tissue and systemic toxicity. Group A streptococci (GAS) and Staphylococcus aureus are two major pathogens associated with monomicrobial NSTIs. In the tissue environment, both Gram-positive bacteria secrete a variety of molecules, including pore-forming exotoxins, superantigens, and proteases with cytolytic and immunomodulatory functions. The present review summarizes the current knowledge about streptococcal and staphylococcal toxins in NSTIs with a special focus on their contribution to disease progression, tissue pathology, and immune evasion strategies.
Collapse
|
11
|
Tam K, Torres VJ. Staphylococcus aureus Secreted Toxins and Extracellular Enzymes. Microbiol Spectr 2019; 7:10.1128/microbiolspec.GPP3-0039-2018. [PMID: 30873936 PMCID: PMC6422052 DOI: 10.1128/microbiolspec.gpp3-0039-2018] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus is a formidable pathogen capable of causing infections in different sites of the body in a variety of vertebrate animals, including humans and livestock. A major contribution to the success of S. aureus as a pathogen is the plethora of virulence factors that manipulate the host's innate and adaptive immune responses. Many of these immune modulating virulence factors are secreted toxins, cofactors for activating host zymogens, and exoenzymes. Secreted toxins such as pore-forming toxins and superantigens are highly inflammatory and can cause leukocyte cell death by cytolysis and clonal deletion, respectively. Coagulases and staphylokinases are cofactors that hijack the host's coagulation system. Exoenzymes, including nucleases and proteases, cleave and inactivate various immune defense and surveillance molecules, such as complement factors, antimicrobial peptides, and surface receptors that are important for leukocyte chemotaxis. Additionally, some of these secreted toxins and exoenzymes can cause disruption of endothelial and epithelial barriers through cell lysis and cleavage of junction proteins. A unique feature when examining the repertoire of S. aureus secreted virulence factors is the apparent functional redundancy exhibited by the majority of the toxins and exoenzymes. However, closer examination of each virulence factor revealed that each has unique properties that have important functional consequences. This chapter provides a brief overview of our current understanding of the major secreted virulence factors critical for S. aureus pathogenesis.
Collapse
Affiliation(s)
- Kayan Tam
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, New York, NY 10016
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, New York, NY 10016
| |
Collapse
|
12
|
Thomsen IP, Liu GY. Targeting fundamental pathways to disrupt Staphylococcus aureus survival: clinical implications of recent discoveries. JCI Insight 2018. [PMID: 29515041 DOI: 10.1172/jci.insight.98216] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The emergence of community-associated methicillin-resistant Staphylococcus aureus during the past decade along with an impending shortage of effective antistaphylococcal antibiotics have fueled impressive advances in our understanding of how S. aureus overcomes the host environment to establish infection. Backed by recent technologic advances, studies have uncovered elaborate metabolic, nutritional, and virulence strategies deployed by S. aureus to survive the restrictive and hostile environment imposed by the host, leading to a plethora of promising antimicrobial approaches that have potential to remedy the antibiotic resistance crisis. In this Review, we highlight some of the critical and recently elucidated bacterial strategies that are potentially amenable to intervention, discuss their relevance to human diseases, and address the translational challenges posed by current animal models.
Collapse
Affiliation(s)
- Isaac P Thomsen
- Department of Pediatrics, Division of Pediatric Infectious Diseases, and Vanderbilt Vaccine Research Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - George Y Liu
- Division of Pediatric Infectious Diseases and Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
13
|
Differential Ability of Pandemic and Seasonal H1N1 Influenza A Viruses To Alter the Function of Human Neutrophils. mSphere 2018; 3:mSphere00567-17. [PMID: 29299535 PMCID: PMC5750393 DOI: 10.1128/mspheredirect.00567-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
A long-standing notion is that IAV inhibits normal neutrophil function and thereby predisposes individuals to secondary bacterial infections. Here we report that seasonal H1N1 IAV primes human neutrophils for enhanced killing of Staphylococcus aureus. Moreover, we provide a comprehensive view of the changes in neutrophil gene expression during interaction with seasonal or pandemic IAV and report how these changes relate to functions such as bactericidal activity. This study expands our knowledge of IAV interactions with human neutrophils. Neutrophils are essential cells of host innate immunity. Although the role of neutrophils in defense against bacterial and fungal infections is well characterized, there is a relative paucity of information about their role against viral infections. Influenza A virus (IAV) infection can be associated with secondary bacterial coinfection, and it has long been posited that the ability of IAV to alter normal neutrophil function predisposes individuals to secondary bacterial infections. To better understand this phenomenon, we evaluated the interaction of pandemic or seasonal H1N1 IAV with human neutrophils isolated from healthy persons. These viruses were ingested by human neutrophils and elicited changes in neutrophil gene expression that are consistent with an interferon-mediated immune response. The viability of neutrophils following coculture with either pandemic or seasonal H1N1 IAV was similar for up to 18 h of culture. Notably, neutrophil exposure to seasonal (but not pandemic) IAV primed these leukocytes for enhanced functions, including production of reactive oxygen species and bactericidal activity. Taken together, our results are at variance with the universal idea that IAV impairs neutrophil function directly to predispose individuals to secondary bacterial infections. Rather, we suggest that some strains of IAV prime neutrophils for enhanced bacterial clearance. IMPORTANCE A long-standing notion is that IAV inhibits normal neutrophil function and thereby predisposes individuals to secondary bacterial infections. Here we report that seasonal H1N1 IAV primes human neutrophils for enhanced killing of Staphylococcus aureus. Moreover, we provide a comprehensive view of the changes in neutrophil gene expression during interaction with seasonal or pandemic IAV and report how these changes relate to functions such as bactericidal activity. This study expands our knowledge of IAV interactions with human neutrophils.
Collapse
|
14
|
Buvelot H, Posfay-Barbe KM, Linder P, Schrenzel J, Krause KH. Staphylococcus aureus, phagocyte NADPH oxidase and chronic granulomatous disease. FEMS Microbiol Rev 2017; 41:139-157. [PMID: 27965320 DOI: 10.1093/femsre/fuw042] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2016] [Indexed: 11/14/2022] Open
Abstract
Dysfunction of phagocytes is a relevant risk factor for staphylococcal infection. The most common hereditary phagocyte dysfunction is chronic granulomatous disease (CGD), characterized by impaired generation of reactive oxygen species (ROS) due to loss of function mutations within the phagocyte NADPH oxidase NOX2. Phagocytes ROS generation is fundamental to eliminate pathogens and to regulate the inflammatory response to infection. CGD is characterized by recurrent and severe bacterial and fungal infections, with Staphylococcus aureus as the most frequent pathogen, and skin and lung abscesses as the most common clinical entities. Staphylococcus aureus infection may occur in virtually any human host, presumably because of the many virulence factors of the bacterium. However, in the presence of functional NOX2, staphylococcal infections remain rare and are mainly linked to breaches of the skin barrier. In contrast, in patients with CGD, S. aureus readily survives and frequently causes clinically apparent disease. Astonishingly, little is known why S. aureus, which possesses a wide range of antioxidant enzymes (e.g. catalase, SOD), is particularly sensitive to control through NOX2. In this review, we will evaluate the discovery of CGD and our present knowledge of the role of NOX2 in S. aureus infection.
Collapse
Affiliation(s)
- Helene Buvelot
- Division of General Internal Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland
| | - Klara M Posfay-Barbe
- Paediatric Infectious Diseases Unit, Department of Paediatrics, University Hospitals of Geneva, 1205 Geneva and Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Patrick Linder
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Jacques Schrenzel
- Divisions of Infectious Diseases and Laboratory Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland
| | - Karl-Heinz Krause
- Divisions of Infectious Diseases and Laboratory Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| |
Collapse
|
15
|
Guerra FE, Borgogna TR, Patel DM, Sward EW, Voyich JM. Epic Immune Battles of History: Neutrophils vs. Staphylococcus aureus. Front Cell Infect Microbiol 2017; 7:286. [PMID: 28713774 PMCID: PMC5491559 DOI: 10.3389/fcimb.2017.00286] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/12/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and the first line of defense after bacteria have breached the epithelial barriers. After migration to a site of infection, neutrophils engage and expose invading microorganisms to antimicrobial peptides and proteins, as well as reactive oxygen species, as part of their bactericidal arsenal. Ideally, neutrophils ingest bacteria to prevent damage to surrounding cells and tissues, kill invading microorganisms with antimicrobial mechanisms, undergo programmed cell death to minimize inflammation, and are cleared away by macrophages. Staphylococcus aureus (S. aureus) is a prevalent Gram-positive bacterium that is a common commensal and causes a wide range of diseases from skin infections to endocarditis. Since its discovery, S. aureus has been a formidable neutrophil foe that has challenged the efficacy of this professional assassin. Indeed, proper clearance of S. aureus by neutrophils is essential to positive infection outcome, and S. aureus has developed mechanisms to evade neutrophil killing. Herein, we will review mechanisms used by S. aureus to modulate and evade neutrophil bactericidal mechanisms including priming, activation, chemotaxis, production of reactive oxygen species, and resolution of infection. We will also highlight how S. aureus uses sensory/regulatory systems to tailor production of virulence factors specifically to the triggering signal, e.g., neutrophils and defensins. To conclude, we will provide an overview of therapeutic approaches that may potentially enhance neutrophil antimicrobial functions.
Collapse
Affiliation(s)
- Fermin E Guerra
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Timothy R Borgogna
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Delisha M Patel
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Eli W Sward
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| |
Collapse
|
16
|
Seilie ES, Bubeck Wardenburg J. Staphylococcus aureus pore-forming toxins: The interface of pathogen and host complexity. Semin Cell Dev Biol 2017; 72:101-116. [PMID: 28445785 DOI: 10.1016/j.semcdb.2017.04.003] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/22/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022]
Abstract
Staphylococcus aureus is a prominent human pathogen capable of infecting a variety of host species and tissue sites. This versatility stems from the pathogen's ability to secrete diverse host-damaging virulence factors. Among these factors, the S. aureus pore-forming toxins (PFTs) α-toxin and the bicomponent leukocidins, have garnered much attention for their ability to lyse cells at low concentrations and modulate disease severity. Although many of these toxins were discovered nearly a century ago, their host cell specificities have only been elucidated over the past five to six years, starting with the discovery of the eukaryotic receptor for α-toxin and rapidly followed by identification of the leukocidin receptors. The identification of these receptors has revealed the species- and cell type-specificity of toxin binding, and provided insight into non-lytic effects of PFT intoxication that contribute to disease pathogenesis.
Collapse
Affiliation(s)
- E Sachiko Seilie
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, United States; Department of Microbiology, The University of Chicago, Chicago, IL 60637, United States
| | | |
Collapse
|
17
|
Spaan AN, van Strijp JAG, Torres VJ. Leukocidins: staphylococcal bi-component pore-forming toxins find their receptors. Nat Rev Microbiol 2017; 15:435-447. [PMID: 28420883 DOI: 10.1038/nrmicro.2017.27] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Staphylococcus aureus is a major bacterial pathogen that causes disease worldwide. The emergence of strains that are resistant to commonly used antibiotics and the failure of vaccine development have resulted in a renewed interest in the pathophysiology of this bacterium. Staphylococcal leukocidins are a family of bi-component pore-forming toxins that are important virulence factors. During the past five years, cellular receptors have been identified for all of the bi-component leukocidins. The identification of the leukocidin receptors explains the cellular tropism and species specificity that is exhibited by these toxins, which has important biological consequences. In this Review, we summarize the recent discoveries that have reignited interest in these toxins and provide an outlook for future research.
Collapse
Affiliation(s)
- András N Spaan
- Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, 10016 New York, USA
| |
Collapse
|
18
|
Zimmermann-Meisse G, Prévost G, Jover E. Above and beyond C5a Receptor Targeting by Staphylococcal Leucotoxins: Retrograde Transport of Panton-Valentine Leucocidin and γ-Hemolysin. Toxins (Basel) 2017; 9:toxins9010041. [PMID: 28117704 PMCID: PMC5308273 DOI: 10.3390/toxins9010041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 12/03/2022] Open
Abstract
Various membrane receptors associated with the innate immune response have recently been identified as mediators of the cellular action of Staphylococcus aureus leucotoxins. Two of these, the Panton–Valentine leucotoxin LukS-PV/LukF-PV and the γ-hemolysin HlgC/HlgB, bind the C5a complement-derived peptide receptor. These leucotoxins utilize the receptor to induce intracellular Ca2+ release from internal stores, other than those activated by C5a. The two leucotoxins are internalized with the phosphorylated receptor, but it is unknown whether they divert retrograde transport of the receptor or follow another pathway. Immunolabeling and confocal microscopic techniques were used to analyze the presence of leucotoxins in endosomes, lysosomes, endoplasmic reticulum, and Golgi. The two leucotoxins apparently followed retrograde transport similar to that of the C5a peptide-activated receptor. However, HlgC/HlgB reached the Golgi network very early, whereas LukS-PV/LukF-PV followed slower kinetics. The HlgC/HlgB leucotoxin remained in neutrophils 6 h after a 10-min incubation of the cells in the presence of the toxin with no signs of apoptosis, whereas apoptosis was observed 3 h after neutrophils were incubated with LukS-PV/LukF-PV. Such retrograde transport of leucotoxins provides a novel understanding of the cellular effects initiated by sublytic concentrations of these toxins.
Collapse
Affiliation(s)
- Gaëlle Zimmermann-Meisse
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), VBP EA7290, Institut de Bactériologie, Université de Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France.
| | - Gilles Prévost
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), VBP EA7290, Institut de Bactériologie, Université de Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France.
| | - Emmanuel Jover
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), VBP EA7290, Institut de Bactériologie, Université de Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France.
| |
Collapse
|
19
|
Is LukS-PV a novel experimental therapy for leukemia? Gene 2016; 600:44-47. [PMID: 27916717 DOI: 10.1016/j.gene.2016.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/17/2016] [Accepted: 11/30/2016] [Indexed: 12/15/2022]
Abstract
Although the studies on the pathogenesis and prognosis of leukemia have made revolutionary progress, the long-term survival remains unsatisfactory. Alternative techniques are being developed to target leukemia. Several decades after researchers' work, a variety of bacteria toxins are being explored as potential anti-leukemia agents, either to provide direct effects or to deliver therapeutic proteins to leukemia. LukS-PV, a component of Panton-Valentine Leukocidin secreted by S. aureus, has been tested in acute myeloid leukemia as a novel experimental strategy. Further researches about the targeting mechanisms of LukS-PV are required to make it a complete therapeutic approach for leukemia treatment. The function of this article is to provide clinicians and experimentalists with a chronological and comprehensive appraisal of use of LukS-PV as an experimental strategy for leukemia therapy.
Collapse
|
20
|
LukS-PV induces differentiation by activating the ERK signaling pathway and c-JUN/c-FOS in human acute myeloid leukemia cells. Int J Biochem Cell Biol 2016; 76:107-14. [DOI: 10.1016/j.biocel.2016.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/11/2016] [Accepted: 04/15/2016] [Indexed: 11/24/2022]
|
21
|
Reyes-Robles T, Torres VJ. Staphylococcus aureus Pore-Forming Toxins. Curr Top Microbiol Immunol 2016; 409:121-144. [PMID: 27406190 DOI: 10.1007/82_2016_16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a formidable foe equipped with an armamentarium of virulence factors to thwart host defenses and establish a successful infection. Among these virulence factors, S. aureus produces several potent secreted proteins that act as cytotoxins, predominant among them the beta-barrel pore-forming toxins. These toxins play several roles in pathogenesis, including disruption of cellular adherens junctions at epithelial barriers, alteration of intracellular signaling events, modulation of host immune responses, and killing of eukaryotic immune and non-immune cells. This chapter provides an updated overview on the S. aureus beta-barrel pore-forming cytotoxins, the identification of toxin receptors on host cells, and their roles in pathogenesis.
Collapse
Affiliation(s)
- Tamara Reyes-Robles
- Department of Microbiology, Microbial Pathogenesis Program, New York University School of Medicine, 522 First Avenue, Smilow Research Building, Room 1010, New York, NY, 10016, USA
| | - Victor J Torres
- Department of Microbiology, Microbial Pathogenesis Program, New York University School of Medicine, 522 First Avenue, Smilow Research Building, Room 1010, New York, NY, 10016, USA.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Methicillin-resistant strains of the important human pathogen Staphylococcus aureus pose a significant public health threat in the community, as they are easily transmitted, especially prone to cause invasive disease, and infect otherwise healthy individuals. The mechanistic basis for the ability of these organisms to evade the innate immune responses remains incompletely defined. RECENT FINDINGS The success of pathogens such as S. aureus rests, in part, on their capacity to overcome neutrophil-mediated host defense to establish infection and cause human disease. S. aureus has the potential to thwart effective neutrophil chemotaxis, and phagocytosis, and succeeds in evading killing by neutrophils. Furthermore, S. aureus surviving within neutrophils promotes neutrophil cytolysis, with release of host-derived molecules that promote local inflammation. Here, we provide a brief overview of our understanding of the mechanisms by which S. aureus - including methicillin-resistant S. aureus - avoids neutrophil-mediated host defense and causes disease. SUMMARY Understanding the molecular mechanisms by which S. aureus avoids neutrophil-mediated responses and initiates signaling cascades that culminate in neutrophil lysis will provide insights prerequisite to the development of novel targets for treating staphylococcal infections.
Collapse
|
23
|
The bicomponent pore-forming leucocidins of Staphylococcus aureus. Microbiol Mol Biol Rev 2015; 78:199-230. [PMID: 24847020 DOI: 10.1128/mmbr.00055-13] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ability to produce water-soluble proteins with the capacity to oligomerize and form pores within cellular lipid bilayers is a trait conserved among nearly all forms of life, including humans, single-celled eukaryotes, and numerous bacterial species. In bacteria, some of the most notable pore-forming molecules are protein toxins that interact with mammalian cell membranes to promote lysis, deliver effectors, and modulate cellular homeostasis. Of the bacterial species capable of producing pore-forming toxic molecules, the Gram-positive pathogen Staphylococcus aureus is one of the most notorious. S. aureus can produce seven different pore-forming protein toxins, all of which are believed to play a unique role in promoting the ability of the organism to cause disease in humans and other mammals. The most diverse of these pore-forming toxins, in terms of both functional activity and global representation within S. aureus clinical isolates, are the bicomponent leucocidins. From the first description of their activity on host immune cells over 100 years ago to the detailed investigations of their biochemical function today, the leucocidins remain at the forefront of S. aureus pathogenesis research initiatives. Study of their mode of action is of immediate interest in the realm of therapeutic agent design as well as for studies of bacterial pathogenesis. This review provides an updated perspective on our understanding of the S. aureus leucocidins and their function, specificity, and potential as therapeutic targets.
Collapse
|
24
|
Shan W, Bu S, Zhang C, Zhang S, Ding B, Chang W, Dai Y, Shen J, Ma X. LukS-PV, a component of Panton-Valentine leukocidin, exerts potent activity against acute myeloid leukemia in vitro and in vivo. Int J Biochem Cell Biol 2015; 61:20-8. [DOI: 10.1016/j.biocel.2015.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/01/2014] [Accepted: 01/09/2015] [Indexed: 12/25/2022]
|
25
|
Tawk MY, Zimmermann K, Bossu J, Potrich C, Bourcier T, Dalla Serra M, Poulain B, Prévost G, Jover E. Internalization of staphylococcal leukotoxins that bind and divert the
C
5a receptor is required for intracellular
Ca
2+
mobilization by human neutrophils. Cell Microbiol 2015; 17:1241-57. [DOI: 10.1111/cmi.12434] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/26/2015] [Accepted: 03/01/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Mira Y. Tawk
- Fédération de Médecine Translationnelle de Strasbourg EA7290 Virulence Bactérienne Précoce Institut de Bactériologie et Hôpitaux Universitaires de Strasbourg Université de Strasbourg Strasbourg France
| | - Kiran Zimmermann
- Fédération de Médecine Translationnelle de Strasbourg EA7290 Virulence Bactérienne Précoce Institut de Bactériologie et Hôpitaux Universitaires de Strasbourg Université de Strasbourg Strasbourg France
| | - Jean‐Louis Bossu
- INCI – UPR‐CNRS 3212 Physiologie des réseaux de neurones Strasbourg France
| | - Cristina Potrich
- National Research Council of Italy Institute of Biophysics and Bruno Kessler Foundation Trento Italy
| | - Tristan Bourcier
- Fédération de Médecine Translationnelle de Strasbourg EA7290 Virulence Bactérienne Précoce Institut de Bactériologie et Hôpitaux Universitaires de Strasbourg Université de Strasbourg Strasbourg France
| | - Mauro Dalla Serra
- National Research Council of Italy Institute of Biophysics and Bruno Kessler Foundation Trento Italy
| | - Bernard Poulain
- INCI – UPR‐CNRS 3212 Physiologie des réseaux de neurones Strasbourg France
| | - Gilles Prévost
- Fédération de Médecine Translationnelle de Strasbourg EA7290 Virulence Bactérienne Précoce Institut de Bactériologie et Hôpitaux Universitaires de Strasbourg Université de Strasbourg Strasbourg France
| | - Emmanuel Jover
- Fédération de Médecine Translationnelle de Strasbourg EA7290 Virulence Bactérienne Précoce Institut de Bactériologie et Hôpitaux Universitaires de Strasbourg Université de Strasbourg Strasbourg France
| |
Collapse
|
26
|
Malachowa N, Kobayashi SD, Sturdevant DE, Scott DP, DeLeo FR. Insights into the Staphylococcus aureus-host interface: global changes in host and pathogen gene expression in a rabbit skin infection model. PLoS One 2015; 10:e0117713. [PMID: 25719526 PMCID: PMC4342162 DOI: 10.1371/journal.pone.0117713] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/31/2014] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is an important cause of human skin and soft tissue infections (SSTIs) globally. Notably, 80% of all SSTIs are caused by S. aureus, of which ∼63% are abscesses and/or cellulitis. Although progress has been made, our knowledge of the host and pathogen factors that contribute to the pathogenesis of SSTIs is incomplete. To provide a more comprehensive view of this process, we monitored changes in the S. aureus transcriptome and selected host proinflammatory molecules during abscess formation and resolution in a rabbit skin infection model. Within the first 24 h, S. aureus transcripts involved in DNA repair, metabolite transport, and metabolism were up-regulated, suggesting an increase in the machinery encoding molecules involved in replication and cell division. There was also increased expression of genes encoding virulence factors, namely secreted toxins and fibronectin and/or fibrinogen-binding proteins. Of the host genes tested, we found that transcripts encoding IL-8, IL1β, oncostatin M-like, CCR1, CXCR1 (IL8RA), CCL4 (MIP-1β) and CCL3 (MIP1α)-like proteins were among the most highly up-regulated transcripts during S. aureus abscess formation. Our findings provide additional insight into the pathogenesis of S. aureus SSTIs, including a temporal component of the host response. These results serve as a springboard for future studies directed to better understand how/why mild or moderate SSTIs progress to invasive disease.
Collapse
Affiliation(s)
- Natalia Malachowa
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Scott D. Kobayashi
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Daniel E. Sturdevant
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Dana P. Scott
- Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Frank R. DeLeo
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
27
|
Spaan AN, Vrieling M, Wallet P, Badiou C, Reyes-Robles T, Ohneck EA, Benito Y, de Haas CJ, Day CJ, Jennings MP, Lina G, Vandenesch F, van Kessel KP, Torres VJ, van Strijp JA, Henry T. The staphylococcal toxins γ-haemolysin AB and CB differentially target phagocytes by employing specific chemokine receptors. Nat Commun 2014; 5:5438. [PMID: 25384670 PMCID: PMC4228697 DOI: 10.1038/ncomms6438] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/01/2014] [Indexed: 12/20/2022] Open
Abstract
Evasion of the host phagocyte response by Staphylococcus aureus is crucial to successful infection with the pathogen. γ-haemolysin AB and CB (HlgAB, HlgCB) are bicomponent pore-forming toxins present in almost all human S. aureus isolates. Cellular tropism and contribution of the toxins to S. aureus pathophysiology are poorly understood. Here we identify the chemokine receptors CXCR1, CXCR2 and CCR2 as targets for HlgAB, and the complement receptors C5aR and C5L2 as targets for HlgCB. The receptor expression patterns allow the toxins to efficiently and differentially target phagocytic cells. Murine neutrophils are resistant to HlgAB and HlgCB. CCR2 is the sole murine receptor orthologue compatible with γ-haemolysin. In a murine peritonitis model, HlgAB contributes to S. aureus bacteremia in a CCR2-dependent manner. HlgAB-mediated targeting of CCR2(+) cells highlights the involvement of inflammatory macrophages during S. aureus infection. Functional quantification identifies HlgAB and HlgCB as major secreted staphylococcal leukocidins.
Collapse
MESH Headings
- Animals
- Bacterial Proteins/physiology
- Bacterial Toxins
- Cells, Cultured
- Disease Models, Animal
- Female
- Hemolysin Proteins/physiology
- Humans
- Macrophages/microbiology
- Macrophages/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Peritonitis/microbiology
- Peritonitis/pathology
- Peritonitis/physiopathology
- Phagocytes/microbiology
- Phagocytes/pathology
- Receptors, CCR2/deficiency
- Receptors, CCR2/genetics
- Receptors, CCR2/physiology
- Receptors, Chemokine/physiology
- Receptors, Complement/physiology
- Receptors, Interleukin-8A/physiology
- Receptors, Interleukin-8B/physiology
- Staphylococcal Infections/pathology
- Staphylococcal Infections/physiopathology
- Staphylococcus aureus
Collapse
Affiliation(s)
- András N. Spaan
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
| | - Manouk Vrieling
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Pierre Wallet
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
- CNRS, UMR5308, 69007 Lyon, France
| | - Cédric Badiou
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
- CNRS, UMR5308, 69007 Lyon, France
| | - Tamara Reyes-Robles
- Department of Microbiology, New York University School of Medicine, 10016 New York, USA
| | - Elizabeth A. Ohneck
- Department of Microbiology, New York University School of Medicine, 10016 New York, USA
| | - Yvonne Benito
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
- CNRS, UMR5308, 69007 Lyon, France
- Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, 69007 Lyon, France
| | - Carla J.C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Christopher J. Day
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Michael P. Jennings
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Gérard Lina
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
- CNRS, UMR5308, 69007 Lyon, France
- Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, 69007 Lyon, France
| | - François Vandenesch
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
- CNRS, UMR5308, 69007 Lyon, France
- Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, 69007 Lyon, France
| | - Kok P.M. van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, 10016 New York, USA
| | - Jos A.G. van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Thomas Henry
- CIRI, International Center for Infectiology Research, Université Lyon 1 and Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Inserm, U1111, 69007 Lyon, France
| |
Collapse
|
28
|
Cheung GYC, Kretschmer D, Duong AC, Yeh AJ, Ho TV, Chen Y, Joo HS, Kreiswirth BN, Peschel A, Otto M. Production of an attenuated phenol-soluble modulin variant unique to the MRSA clonal complex 30 increases severity of bloodstream infection. PLoS Pathog 2014; 10:e1004298. [PMID: 25144687 PMCID: PMC4140855 DOI: 10.1371/journal.ppat.1004298] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/20/2014] [Indexed: 12/19/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of morbidity and death. Phenol-soluble modulins (PSMs) are recently-discovered toxins with a key impact on the development of Staphylococcus aureus infections. Allelic variants of PSMs and their potential impact on pathogen success during infection have not yet been described. Here we show that the clonal complex (CC) 30 lineage, a major cause of hospital-associated sepsis and hematogenous complications, expresses an allelic variant of the PSMα3 peptide. We found that this variant, PSMα3N22Y, is characteristic of CC30 strains and has significantly reduced cytolytic and pro-inflammatory potential. Notably, CC30 strains showed reduced cytolytic and chemotactic potential toward human neutrophils, and increased hematogenous seeding in a bacteremia model, compared to strains in which the genome was altered to express non-CC30 PSMα3. Our findings describe a molecular mechanism contributing to attenuated pro-inflammatory potential in a main MRSA lineage. They suggest that reduced pathogen recognition via PSMs allows the bacteria to evade elimination by innate host defenses during bloodstream infections. Furthermore, they underscore the role of point mutations in key S. aureus toxin genes in that adaptation and the pivotal importance PSMs have in defining key S. aureus immune evasion and virulence mechanisms. Methicillin-resistant Staphylococcus aureus (MRSA) is a major cause of morbidity and mortality and a great concern for public health. The CC30 MRSA lineage is especially notorious for causing bloodstream infections with complications such as seeding into organs. In our study, we show that this lineage produces an attenuated form of a key S. aureus toxin with decreased pro-inflammatory features. Our results suggest that attenuation of this toxin allows the bacteria to evade recognition and subsequent elimination by host defenses, thereby increasing pathogen success during blood infection.
Collapse
Affiliation(s)
- Gordon Y. C. Cheung
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dorothee Kretschmer
- Cellular and Molecular Microbiology Division, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Anthony C. Duong
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anthony J. Yeh
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Trung V. Ho
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yan Chen
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hwang-Soo Joo
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Barry N. Kreiswirth
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Andreas Peschel
- Cellular and Molecular Microbiology Division, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
29
|
Aman MJ, Adhikari RP. Staphylococcal bicomponent pore-forming toxins: targets for prophylaxis and immunotherapy. Toxins (Basel) 2014; 6:950-72. [PMID: 24599233 PMCID: PMC3968370 DOI: 10.3390/toxins6030950] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/22/2014] [Accepted: 02/26/2014] [Indexed: 01/09/2023] Open
Abstract
Staphylococccus aureus represents one of the most challenging human pathogens as well as a common colonizer of human skin and mucosal surfaces. S. aureus causes a wide range of diseases from skin and soft tissue infection (SSTI) to debilitating and life-threatening conditions such as osteomyelitis, endocarditis, and necrotizing pneumonia. The range of diseases reflects the remarkable diversity of the virulence factors produced by this pathogen, including surface antigens involved in the establishment of infection and a large number of toxins that mediate a vast array of cellular responses. The staphylococcal toxins are generally believed to have evolved to disarm the innate immune system, the first line of defense against this pathogen. This review focuses on recent advances on elucidating the biological functions of S. aureus bicomponent pore-forming toxins (BCPFTs) and their utility as targets for preventive and therapeutic intervention. These toxins are cytolytic to a variety of immune cells, primarily neutrophils, as well as cells with a critical barrier function. The lytic activity of BCPFTs towards immune cells implies a critical role in immune evasion, and a number of epidemiological studies and animal experiments relate these toxins to clinical disease, particularly SSTI and necrotizing pneumonia. Antibody-mediated neutralization of this lytic activity may provide a strategy for development of toxoid-based vaccines or immunotherapeutics for prevention or mitigation of clinical diseases. However, certain BCPFTs have been proposed to act as danger signals that may alert the immune system through an inflammatory response. The utility of a neutralizing vaccination strategy must be weighed against such immune-activating potential.
Collapse
Affiliation(s)
- M Javad Aman
- Integrated BioTherapeutics Inc., 21 Firstfield Rd., Gaithersburg, MD 20878, USA.
| | - Rajan P Adhikari
- Integrated BioTherapeutics Inc., 21 Firstfield Rd., Gaithersburg, MD 20878, USA.
| |
Collapse
|
30
|
Spaan AN, Henry T, van Rooijen WJM, Perret M, Badiou C, Aerts PC, Kemmink J, de Haas CJC, van Kessel KPM, Vandenesch F, Lina G, van Strijp JAG. The staphylococcal toxin Panton-Valentine Leukocidin targets human C5a receptors. Cell Host Microbe 2013; 13:584-594. [PMID: 23684309 DOI: 10.1016/j.chom.2013.04.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 03/15/2013] [Accepted: 04/09/2013] [Indexed: 12/14/2022]
Abstract
Panton-Valentine Leukocidin (PVL) is a staphylococcal bicomponent pore-forming toxin linked to severe invasive infections. Target-cell and species specificity of PVL are poorly understood, and the mechanism of action of this toxin in Staphylococcus aureus virulence is controversial. Here, we identify the human complement receptors C5aR and C5L2 as host targets of PVL, mediating both toxin binding and cytotoxicity. Expression and interspecies variations of the C5aR determine cell and species specificity of PVL. The C5aR binding PVL component, LukS-PV, is a potent inhibitor of C5a-induced immune cell activation. These findings provide insight into leukocidin function and staphylococcal virulence and offer directions for future investigations into individual susceptibility to severe staphylococcal disease.
Collapse
Affiliation(s)
- András N Spaan
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Thomas Henry
- CIRI, International Center for Infectiology Research, LabEx Ecofect, Université Lyon 1, 69007 Lyon, France; Inserm, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; CNRS, UMR5308, 69007 Lyon, France
| | | | - Magali Perret
- CIRI, International Center for Infectiology Research, LabEx Ecofect, Université Lyon 1, 69007 Lyon, France; Inserm, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; CNRS, UMR5308, 69007 Lyon, France
| | - Cédric Badiou
- CIRI, International Center for Infectiology Research, LabEx Ecofect, Université Lyon 1, 69007 Lyon, France; Inserm, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; CNRS, UMR5308, 69007 Lyon, France
| | - Piet C Aerts
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Johan Kemmink
- Medicinal Chemistry and Chemical Biology, Utrecht University, 3584CX Utrecht, The Netherlands
| | - Carla J C de Haas
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Kok P M van Kessel
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - François Vandenesch
- CIRI, International Center for Infectiology Research, LabEx Ecofect, Université Lyon 1, 69007 Lyon, France; Inserm, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; CNRS, UMR5308, 69007 Lyon, France; Hospices Civils de Lyon, 69007 Lyon, France
| | - Gérard Lina
- CIRI, International Center for Infectiology Research, LabEx Ecofect, Université Lyon 1, 69007 Lyon, France; Inserm, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; CNRS, UMR5308, 69007 Lyon, France; Hospices Civils de Lyon, 69007 Lyon, France
| | - Jos A G van Strijp
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands.
| |
Collapse
|
31
|
Malachowa N, Kobayashi SD, Freedman B, Dorward DW, DeLeo FR. Staphylococcus aureus leukotoxin GH promotes formation of neutrophil extracellular traps. THE JOURNAL OF IMMUNOLOGY 2013; 191:6022-9. [PMID: 24190656 DOI: 10.4049/jimmunol.1301821] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus secretes numerous virulence factors that facilitate evasion of the host immune system. Among these molecules are pore-forming cytolytic toxins, including Panton-Valentine leukocidin (PVL), leukotoxin GH (LukGH; also known as LukAB), leukotoxin DE, and γ-hemolysin. PVL and LukGH have potent cytolytic activity in vitro, and both toxins are proinflammatory in vivo. Although progress has been made toward elucidating the role of these toxins in S. aureus virulence, our understanding of the mechanisms that underlie the proinflammatory capacity of these toxins, as well as the associated host response toward them, is incomplete. To address this deficiency in knowledge, we assessed the ability of LukGH to prime human PMNs for enhanced bactericidal activity and further investigated the impact of the toxin on neutrophil function. We found that, unlike PVL, LukGH did not prime human neutrophils for increased production of reactive oxygen species nor did it enhance binding and/or uptake of S. aureus. Unexpectedly, LukGH promoted the release of neutrophil extracellular traps (NETs), which, in turn, ensnared but did not kill S. aureus. Furthermore, we found that electropermeabilization of human neutrophils, used as a separate means to create pores in the neutrophil plasma membrane, similarly induced formation of NETs, a finding consistent with the notion that NETs can form during nonspecific cytolysis. We propose that the ability of LukGH to promote formation of NETs contributes to the inflammatory response and host defense against S. aureus infection.
Collapse
Affiliation(s)
- Natalia Malachowa
- Laboratory of Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | | | | | | | | |
Collapse
|
32
|
Kobayashi SD, Olsen RJ, LaCasse RA, Safronetz D, Ashraf M, Porter AR, Braughton KR, Feldmann F, Clifton DR, Kash JC, Bailey JR, Gardner DJ, Otto M, Brining DL, Kreiswirth BN, Taubenberger JK, Parnell MJ, Feldmann H, Musser JM, DeLeo FR. Seasonal H3N2 influenza A virus fails to enhance Staphylococcus aureus co-infection in a non-human primate respiratory tract infection model. Virulence 2013; 4:707-15. [PMID: 24104465 DOI: 10.4161/viru.26572] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Staphylococcus aureus community-acquired pneumonia is often associated with influenza or an influenza-like syndrome. Morbidity and mortality due to methicillin-resistant S. aureus (MRSA) or influenza and pneumonia, which includes bacterial co-infection, are among the top causes of death by infectious diseases in the United States. We developed a non-lethal influenza A virus (IAV) (H3N2)/S. aureus co-infection model in cynomolgus macaques (Macaca fascicularis) to test the hypothesis that seasonal IAV infection predisposes non-human primates to severe S. aureus pneumonia. Infection and disease progression were monitored by clinical assessment of animal health; analysis of blood chemistry, nasal swabs, and X-rays; and gross pathology and histopathology of lungs from infected animals. Seasonal IAV infection in healthy cynomolgus macaques caused mild pneumonia, but unexpectedly, did not predispose these animals to subsequent severe infection with the community-associated MRSA clone USA300. We conclude that in our co-infection model, seasonal IAV infection alone is not sufficient to promote severe S. aureus pneumonia in otherwise healthy non-human primates. The implication of these findings is that comorbidity factors in addition to IAV infection are required to predispose individuals to secondary S. aureus pneumonia.
Collapse
Affiliation(s)
- Scott D Kobayashi
- Laboratory of Human Bacterial Pathogenesis; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Randall J Olsen
- Center for Molecular and Translational Human Infectious Disease Research; The Methodist Hospital Research Institute; Houston, TX USA; Department of Pathology and Genomic Medicine; The Methodist Hospital; Houston, TX USA
| | - Rachel A LaCasse
- Veterinary Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - David Safronetz
- Laboratory of Virology; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Madiha Ashraf
- Center for Molecular and Translational Human Infectious Disease Research; The Methodist Hospital Research Institute; Houston, TX USA; Department of Medicine; The Methodist Hospital; Houston, TX USA
| | - Adeline R Porter
- Laboratory of Human Bacterial Pathogenesis; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Kevin R Braughton
- Laboratory of Human Bacterial Pathogenesis; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Friederike Feldmann
- Office of Operations Management; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Dawn R Clifton
- Laboratory of Virology; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - John C Kash
- Laboratory of Infectious Diseases; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Bethesda, MD USA
| | - John R Bailey
- Veterinary Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Donald J Gardner
- Veterinary Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Michael Otto
- Laboratory of Human Bacterial Pathogenesis; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Douglas L Brining
- Veterinary Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Barry N Kreiswirth
- Public Health Research Institute; University of Medicine and Dentistry of New Jersey; Newark, NJ USA
| | - Jeffrey K Taubenberger
- Laboratory of Infectious Diseases; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Bethesda, MD USA
| | - Michael J Parnell
- Veterinary Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - Heinz Feldmann
- Laboratory of Virology; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| | - James M Musser
- Center for Molecular and Translational Human Infectious Disease Research; The Methodist Hospital Research Institute; Houston, TX USA; Department of Pathology and Genomic Medicine; The Methodist Hospital; Houston, TX USA
| | - Frank R DeLeo
- Laboratory of Human Bacterial Pathogenesis; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Hamilton, MT USA
| |
Collapse
|
33
|
Yoong P, Torres VJ. The effects of Staphylococcus aureus leukotoxins on the host: cell lysis and beyond. Curr Opin Microbiol 2013; 16:63-9. [PMID: 23466211 DOI: 10.1016/j.mib.2013.01.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/03/2012] [Accepted: 01/28/2013] [Indexed: 12/24/2022]
Abstract
The success of Staphylococcus aureus as a leading cause of deadly hospital-acquired and community-acquired infections is attributed to its high-level resistance to most antibiotics, and the multitude of virulence factors it elaborates. Most clinical isolates produce up to four bi-component pore-forming toxins capable of lysing cells of the immune system. Subtle differences in activity and target range of each leukotoxin suggest that these toxins are not redundant, but instead may have specialized functions in attacking and/or evading host defenses. In turn, the host has developed countermeasures recognizing sublytic levels of leukotoxins as signals to activate protective immune defenses. The opposing cytotoxic and immune-activating effects of leukotoxins on host cells make for a complex dynamic between S. aureus and the host.
Collapse
Affiliation(s)
- Pauline Yoong
- Department of Microbiology, New York University School of Medicine, United States
| | | |
Collapse
|
34
|
Staphylococcus aureus LukAB cytotoxin kills human neutrophils by targeting the CD11b subunit of the integrin Mac-1. Proc Natl Acad Sci U S A 2013; 110:10794-9. [PMID: 23754403 DOI: 10.1073/pnas.1305121110] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus causes diseases ranging from superficial wound infections to more invasive manifestations like osteomyelitis and endocarditis. The evasion of host phagocytes recruited to the site of infection is essential to the success of S. aureus as a pathogen. A single S. aureus strain can produce up to five different bicomponent pore-forming leukotoxins that lyse immune cells by forming pores in the cellular plasma membrane. Although these leukotoxins have been considered redundant due to their cytotoxic activity toward human neutrophils, each toxin displays varied species and cell-type specificities. This suggests that cellular factors may influence which cells each toxin targets. Here we describe the identification of CD11b, the α subunit of the αM/β2 integrin (CD11b/CD18), macrophage-1 antigen, or complement receptor 3, as a cellular receptor for leukocidin A/B (LukAB), an important toxin that contributes to S. aureus killing of human neutrophils. We demonstrate that CD11b renders human neutrophils susceptible to LukAB-mediated killing by purified LukAB as well as during S. aureus infection ex vivo. LukAB directly interacts with human CD11b by binding to the I domain, a property that determines the species specificity exhibited by this toxin. Identification of a LukAB cellular target has broad implications for the use of animal models to study the role of LukAB in S. aureus pathogenesis, explains the toxin's tropism toward human neutrophils and other phagocytes, and provides a cellular therapeutic target to block the effect of LukAB toward human neutrophils.
Collapse
|
35
|
Uhlemann AC, Kennedy AD, Martens C, Porcella SF, Deleo FR, Lowy FD. Toward an understanding of the evolution of Staphylococcus aureus strain USA300 during colonization in community households. Genome Biol Evol 2013; 4:1275-85. [PMID: 23104992 PMCID: PMC3542572 DOI: 10.1093/gbe/evs094] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is a frequent cause of serious infections and also a human commensal. The emergence of community-associated methicillin-resistant S. aureus led to a dramatic increase in skin and soft tissue infections worldwide. This epidemic has been driven by a limited number of clones, such as USA300 in the United States. To better understand the extent of USA300 evolution and diversification within communities, we performed comparative whole-genome sequencing of three clinical and five colonizing USA300 isolates collected longitudinally from three unrelated households over a 15-month period. Phylogenetic analysis that incorporated additional geographically diverse USA300 isolates indicated that all but one likely arose from a common recent ancestor. Although limited genetic adaptation occurred over the study period, the greatest genetic heterogeneity occurred between isolates from different households and within one heavily colonized household. This diversity allowed for a more accurate tracking of interpersonal USA300 transmission. Sequencing of persisting USA300 isolates revealed mutations in genes involved in major aspects of S. aureus function: adhesion, cell wall biosynthesis, virulence, and carbohydrate metabolism. Genetic variations also included accumulation of multiple polymorphisms within select genes of two multigene operons, suggestive of small genome rearrangements rather than de novo single point mutations. Such rearrangements have been underappreciated in S. aureus and may represent novel means of strain variation. Subtle genetic changes may contribute to USA300 fitness and persistence. Elucidation of small genome rearrangements reveals a potentially new and intriguing mechanism of directed S. aureus genome diversification in environmental niches and during pathogen-host interactions.
Collapse
Affiliation(s)
- Anne-Catrin Uhlemann
- Division of Infectious Diseases, Department of Medicine, Columbia University, College of Physicians & Surgeons, New York, NY, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Matsuo R, Ago T, Kamouchi M, Kuroda J, Kuwashiro T, Hata J, Sugimori H, Fukuda K, Gotoh S, Makihara N, Fukuhara M, Awano H, Isomura T, Suzuki K, Yasaka M, Okada Y, Kiyohara Y, Kitazono T. Clinical significance of plasma VEGF value in ischemic stroke - research for biomarkers in ischemic stroke (REBIOS) study. BMC Neurol 2013; 13:32. [PMID: 23566234 PMCID: PMC3637234 DOI: 10.1186/1471-2377-13-32] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 03/27/2013] [Indexed: 12/31/2022] Open
Abstract
Background Vascular endothelial growth factor (VEGF) is a well-known molecule mediating neuronal survival and angiogenesis. However, its clinical significance in ischemic stroke is still controversial. The goal of this study was to examine the temporal profile of plasma VEGF value and its clinical significance in ischemic stroke with taking its subtypes into consideration. Methods We prospectively enrolled 171 patients with ischemic stroke and age- and gender-matched healthy subjects. The stroke patients were divided into 4 subtypes: atherothrombotic infarction (ATBI, n = 34), lacunar infarction (LAC, n = 45), cardioembolic infarction (CE, n = 49) and other types (OT, n = 43). Plasma VEGF values were measured as a part of multiplex immunoassay (Human MAP v1.6) and we obtained clinical information at 5 time points (days 0, 3, 7, 14 and 90) after the stroke onset. Results Plasma VEGF values were significantly higher in all stroke subtypes but OT than those in the controls throughout 90 days after stroke onset. There was no significant difference in the average VEGF values among ATBI, LAC, and CE. VEGF values were positively associated with neurological severity in CE patients, while a negative association was found in ATBI patients. After adjustment for possible confounding factors, plasma VEGF value was an independent predictor of poor functional outcome in CE patients. Conclusions Although plasma VEGF value increases immediately after the stroke onset equally in all stroke subtypes, its significance in functional outcome may be different among the stroke subtypes.
Collapse
Affiliation(s)
- Ryu Matsuo
- Department of Medicine and Clinical Science, Kyushu University, 3-1-1 Maidashi, higashi-ku, Fukuoka, 812-8582, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Prabhakara S, Khedkar S, Shambat SM, Srinivasan R, Basu A, Norrby-Teglund A, Seshasayee ASN, Arakere G. Genome sequencing unveils a novel sea enterotoxin-carrying PVL phage in Staphylococcus aureus ST772 from India. PLoS One 2013; 8:e60013. [PMID: 23544121 PMCID: PMC3609733 DOI: 10.1371/journal.pone.0060013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 02/20/2013] [Indexed: 12/26/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen, first recognized as a leading cause of hospital-acquired infections. Community-associated S. aureus (CA-SA) pose a greater threat due to increase in severity of infection and disease among children and healthy adults. CA-SA strains in India are genetically diverse, among which is the sequence type (ST) 772, which has now spread to Australia, Europe and Japan. Towards understanding the genetic characteristics of ST772, we obtained draft genome sequences of five relevant clinical isolates and studied the properties of their PVL-carrying prophages, whose presence is a defining hallmark of CA-SA. We show that this is a novel prophage, which carries the structural genes of the hlb-carrying prophage and includes the sea enterotoxin. This architecture probably emerged early within the ST772 lineage, at least in India. The sea gene, unique to ST772 PVL, despite having promoter sequence characteristics typical of low expression, appears to be highly expressed during early phase of growth in laboratory conditions. We speculate that this might be a consequence of its novel sequence context. The crippled nature of the hlb-converting prophage in ST772 suggests that widespread mobility of the sea enterotoxin might be a selective force behind its 'transfer' to the PVL prophage. Wild type ST772 strains induced strong proliferative responses as well as high cytotoxic activity against neutrophils, likely mediated by superantigen SEA and the PVL toxin respectively. Both proliferation and cytotoxicity were markedly reduced in a cured ST772 strain indicating the impact of the phage on virulence. The presence of SEA alongside the genes for the immune system-modulating PVL toxin may contribute to the success and virulence of ST772.
Collapse
Affiliation(s)
- Sushma Prabhakara
- Society for Innovation and Development, Indian Institute of Science, Bengaluru, India
| | - Supriya Khedkar
- National Centre for Biological Sciences, TIFR, GKVK, Bengaluru, India
| | - Srikanth Mairpady Shambat
- Karolinska Institute, Center for Infectious Medicine F59, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | | - Atanu Basu
- National Institute of Virology, Pune, India
| | - Anna Norrby-Teglund
- Karolinska Institute, Center for Infectious Medicine F59, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | | - Gayathri Arakere
- Society for Innovation and Development, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
38
|
Bacterial survival amidst an immune onslaught: the contribution of the Staphylococcus aureus leukotoxins. PLoS Pathog 2013; 9:e1003143. [PMID: 23436994 PMCID: PMC3578777 DOI: 10.1371/journal.ppat.1003143] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
39
|
Jover E, Tawk MY, Laventie BJ, Poulain B, Prévost G. Staphylococcal leukotoxins trigger free intracellular Ca(2+) rise in neurones, signalling through acidic stores and activation of store-operated channels. Cell Microbiol 2012; 15:742-58. [PMID: 23152983 PMCID: PMC3654557 DOI: 10.1111/cmi.12069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 10/18/2012] [Accepted: 11/06/2012] [Indexed: 12/31/2022]
Abstract
Headache, muscle aches and chest pain of mild to medium intensity are among the most common clinical symptoms in moderate Staphylococcus aureus infections, with severe infections usually associated with worsening pain symptoms. These nociceptive responses of the body raise the question of how bacterial infection impinges on the nervous system. Does S. aureus, or its released virulence factors, act directly on neurones? To address this issue, we evaluated the potential effects on neurones of certain bi-component leukotoxins, which are virulent factors released by the bacterium. The activity of four different leukotoxins was verified by measuring the release of glutamate from rat cerebellar granular neurones. The bi-component γ-haemolysin HlgC/HlgB was the most potent leukotoxin, initiating transient rises in intracellular Ca2+ concentration in cerebellar neurones and in primary sensory neurones from dorsal root ganglia, as probed with the Fura-2 Ca2+ indicator dye. Using pharmacological antagonists of receptors and Ca2+ channels, the variations in intracellular Ca2+ concentration were found independent of the activation of voltage-operatedCa2+ channels or glutamate receptors. Drugs targeting Sarco-Endoplasmic Reticulum Ca2+-ATPase (SERCA) or H+-ATPase and antagonists of the store-operated Ca2+ entry complex blunted, or significantly reduced, the leukotoxin-induced elevation in intracellular Ca2+. Moreover, activation of the ADP-ribosyl cyclase CD38 was also required to initiate the release of Ca2+ from acidic stores. These findings suggest that, prior to forming a pore at the plasma membrane, leukotoxin HlgC/HlgB triggers a multistep process which initiates the release of Ca2+ from lysosomes, modifies the steady-state level of reticular Ca2+ stores and finally activates the Store-Operated Calcium Entry complex.
Collapse
Affiliation(s)
- Emmanuel Jover
- INCI - UPR-CNRS 3212, Neurotransmission et sécrétion neuroendocrine, 5, rue Blaise Pascal, F- 67084 Strasbourg cedex, France. jover@inci-cnrs
| | | | | | | | | |
Collapse
|
40
|
Malachowa N, Kobayashi SD, Braughton KR, Whitney AR, Parnell MJ, Gardner DJ, Deleo FR. Staphylococcus aureus leukotoxin GH promotes inflammation. J Infect Dis 2012; 206:1185-93. [PMID: 22872735 DOI: 10.1093/infdis/jis495] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Staphylococcus aureus produces numerous molecules that facilitate survival in the host. We recently identified a novel S. aureus leukotoxin (leukotoxin GH [LukGH]) using proteomics, but its role in virulence remains unclear. Here we investigated the role of LukGH in vivo. METHODS We tested cytotoxicity of LukGH toward polymorphonuclear leukocytes (PMNs) from mice, rabbits, monkeys, and humans. LukGH was administered to mice, rabbits, and a cynomolgus monkey by subcutaneous or intradermal injection to assess cytotoxicity or host response in vivo. The effects of LukGH in vivo were compared with those of Panton-Valentine leukocidin (PVL), a well-characterized S. aureus leukotoxin. The contribution of LukGH to S. aureus infection was tested using mouse and rabbit infection models. RESULTS Susceptibility of PMNs to LukGH was similar between humans and cynomolgus monkeys, and was greater than that of rabbits, which in turn was greater than that of mice. LukGH or PVL caused skin inflammation in rabbits and a monkey, but deletion of neither lukGH nor lukGH and lukS/F-PV reduced severity of USA300 infections in rabbits or mice. Rather, some disease parameters (eg, rabbit abscess size) were increased following infection with a lukGH and lukS/F-PV deletion strain. CONCLUSIONS Our findings indicate that S. aureus leukotoxins enhance the host inflammatory response and influence the outcome of infection.
Collapse
Affiliation(s)
- Natalia Malachowa
- Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | | | | | | | | | | | | |
Collapse
|