1
|
Lubkin A, Bernard-Raichon L, DuMont AL, Valero Jimenez AM, Putzel GG, Gago J, Zwack EE, Olusanya O, Boguslawski KM, Dallari S, Dyzenhaus S, Herrmann C, Ilmain JK, Isom GL, Pawline M, Perault AI, Perelman S, Sause WE, Shahi I, St. John A, Tierce R, Zheng X, Zhou C, Noval MG, O'Keeffe A, Podkowik M, Gonzales S, Inglima K, Desvignes L, Hochman SE, Stapleford KA, Thorpe LE, Pironti A, Shopsin B, Cadwell K, Dittmann M, Torres VJ. SARS-CoV-2 infection predisposes patients to coinfection with Staphylococcus aureus. mBio 2024; 15:e0166724. [PMID: 39037272 PMCID: PMC11323729 DOI: 10.1128/mbio.01667-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024] Open
Abstract
Severe COVID-19 has been associated with coinfections with bacterial and fungal pathogens. Notably, patients with COVID-19 who develop Staphylococcus aureus bacteremia exhibit higher rates of mortality than those infected with either pathogen alone. To understand this clinical scenario, we collected and examined S. aureus blood and respiratory isolates from a hospital in New York City during the early phase of the pandemic from both SARS-CoV-2+ and SARS-CoV-2- patients. Whole genome sequencing of these S. aureus isolates revealed broad phylogenetic diversity in both patient groups, suggesting that SARS-CoV-2 coinfection was not associated with a particular S. aureus lineage. Phenotypic characterization of the contemporary collection of S. aureus isolates from SARS-CoV-2+ and SARS-CoV-2- patients revealed no notable differences in several virulence traits examined. However, we noted a trend toward overrepresentation of S. aureus bloodstream strains with low cytotoxicity in the SARS-CoV-2+ group. We observed that patients coinfected with SARS-CoV-2 and S. aureus were more likely to die during the acute phase of infection when the coinfecting S. aureus strain exhibited high or low cytotoxicity. To further investigate the relationship between SARS-CoV-2 and S. aureus infections, we developed a murine coinfection model. These studies revealed that infection with SARS-CoV-2 renders mice susceptible to subsequent superinfection with low cytotoxicity S. aureus. Thus, SARS-CoV-2 infection sensitizes the host to coinfections, including S. aureus isolates with low intrinsic virulence. IMPORTANCE The COVID-19 pandemic has had an enormous impact on healthcare across the globe. Patients who were severely infected with SARS-CoV-2, the virus causing COVID-19, sometimes became infected with other pathogens, which is termed coinfection. If the coinfecting pathogen is the bacterium Staphylococcus aureus, there is an increased risk of patient death. We collected S. aureus strains that coinfected patients with SARS-CoV-2 to study the disease outcome caused by the interaction of these two important pathogens. We found that both in patients and in mice, coinfection with an S. aureus strain lacking toxicity resulted in more severe disease during the early phase of infection, compared with infection with either pathogen alone. Thus, SARS-CoV-2 infection can directly increase the severity of S. aureus infection.
Collapse
Affiliation(s)
- Ashira Lubkin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Lucie Bernard-Raichon
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ashley L. DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ana Mayela Valero Jimenez
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Gregory G. Putzel
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
| | - Juan Gago
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| | - Erin E. Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Olufolakemi Olusanya
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
| | - Kristina M. Boguslawski
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Simone Dallari
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sophie Dyzenhaus
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Christin Herrmann
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Juliana K. Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Georgia L. Isom
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Miranda Pawline
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Andrew I. Perault
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
| | - Sofya Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - William E. Sause
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ifrah Shahi
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Amelia St. John
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Rebecca Tierce
- Division of Comparative Medicine, New York University Langone Health, New York, New York, USA
| | - Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Chunyi Zhou
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Maria G. Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Anna O'Keeffe
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Magda Podkowik
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
| | - Sandra Gonzales
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Kenneth Inglima
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Ludovic Desvignes
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
- High Containment Laboratories, Office of Science and Research, NYU Langone Health, New York, New York, USA
| | - Sarah E. Hochman
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Lorna E. Thorpe
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Meike Dittmann
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, Microbial Genomics Core Lab, New York University Langone Health, New York, New York, USA
| |
Collapse
|
2
|
Lichota A, Gwozdzinski K, Kowalczyk E, Kowalczyk M, Sienkiewicz M. Contribution of staphylococcal virulence factors in the pathogenesis of thrombosis. Microbiol Res 2024; 283:127703. [PMID: 38537329 DOI: 10.1016/j.micres.2024.127703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Staphylococci are responsible for many infections in humans, starting with skin and soft tissue infections and finishing with invasive diseases such as endocarditis, sepsis and pneumonia, which lead to high mortality. Patients with sepsis often demonstrate activated clotting pathways, decreased levels of anticoagulants, decreased fibrinolysis, activated endothelial surfaces and activated platelets. This results in disseminated intravascular coagulation and formation of a microthrombus, which can lead to a multiorgan failure. This review describes various staphylococcal virulence factors that contribute to vascular thrombosis, including deep vein thrombosis in infected patients. The article presents mechanisms of action of different factors released by bacteria in various host defense lines, which in turn can lead to formation of blood clots in the vessels.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland.
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Lodz, Poland
| | | | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
3
|
Lê-Bury P, Echenique-Rivera H, Pizarro-Cerdá J, Dussurget O. Determinants of bacterial survival and proliferation in blood. FEMS Microbiol Rev 2024; 48:fuae013. [PMID: 38734892 PMCID: PMC11163986 DOI: 10.1093/femsre/fuae013] [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: 11/06/2023] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/13/2024] Open
Abstract
Bloodstream infection is a major public health concern associated with high mortality and high healthcare costs worldwide. Bacteremia can trigger fatal sepsis whose prevention, diagnosis, and management have been recognized as a global health priority by the World Health Organization. Additionally, infection control is increasingly threatened by antimicrobial resistance, which is the focus of global action plans in the framework of a One Health response. In-depth knowledge of the infection process is needed to develop efficient preventive and therapeutic measures. The pathogenesis of bloodstream infection is a dynamic process resulting from the invasion of the vascular system by bacteria, which finely regulate their metabolic pathways and virulence factors to overcome the blood immune defenses and proliferate. In this review, we highlight our current understanding of determinants of bacterial survival and proliferation in the bloodstream and discuss their interactions with the molecular and cellular components of blood.
Collapse
Affiliation(s)
- Pierre Lê-Bury
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 18 route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Hebert Echenique-Rivera
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Yersinia National Reference Laboratory, WHO Collaborating Research & Reference Centre for Plague FRA-146, 28 rue du Dr Roux, 75015 Paris, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
4
|
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
|
5
|
Jiang JH, Cameron DR, Nethercott C, Aires-de-Sousa M, Peleg AY. Virulence attributes of successful methicillin-resistant Staphylococcus aureus lineages. Clin Microbiol Rev 2023; 36:e0014822. [PMID: 37982596 PMCID: PMC10732075 DOI: 10.1128/cmr.00148-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of severe and often fatal infections. MRSA epidemics have occurred in waves, whereby a previously successful lineage has been replaced by a more fit and better adapted lineage. Selection pressures in both hospital and community settings are not uniform across the globe, which has resulted in geographically distinct epidemiology. This review focuses on the mechanisms that trigger the establishment and maintenance of current, dominant MRSA lineages across the globe. While the important role of antibiotic resistance will be mentioned throughout, factors which influence the capacity of S. aureus to colonize and cause disease within a host will be the primary focus of this review. We show that while MRSA possesses a diverse arsenal of toxins including alpha-toxin, the success of a lineage involves more than just producing toxins that damage the host. Success is often attributed to the acquisition or loss of genetic elements involved in colonization and niche adaptation such as the arginine catabolic mobile element, as well as the activity of regulatory systems, and shift metabolism accordingly (e.g., the accessory genome regulator, agr). Understanding exactly how specific MRSA clones cause prolonged epidemics may reveal targets for therapies, whereby both core (e.g., the alpha toxin) and acquired virulence factors (e.g., the Panton-Valentine leukocidin) may be nullified using anti-virulence strategies.
Collapse
Affiliation(s)
- Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David R. Cameron
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Cara Nethercott
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marta Aires-de-Sousa
- Laboratory of Molecular Genetics, Institutode Tecnologia Químicae Biológica António Xavier (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
- Escola Superior de Saúde da Cruz Vermelha Portuguesa-Lisboa (ESSCVP-Lisboa), Lisbon, Portugal
| | - Anton Y. Peleg
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Anderson EE, Ilmain JK, Torres VJ. SarS and Rot are necessary for the repression of lukED and lukSF-PV in Staphylococcus aureus. Microbiol Spectr 2023; 11:e0165623. [PMID: 37800956 PMCID: PMC10715151 DOI: 10.1128/spectrum.01656-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE The leukocidins play an important role in disarming the host immune system and promoting infection. While both SarS and Rot have been established as repressors of leukocidins, the importance of each repressor in infection is unclear. Here, we demonstrate that repression by SarS and Rot is not additive and show that in addition to upregulating expression of each other, they are also able to bind concurrently to the leukocidin promoters. These findings suggest that both repressors are necessary for maximal repression of lukED and lukSF-PV and illuminate another complex relationship among Staphylococcus aureus virulence regulators.
Collapse
Affiliation(s)
- Exene E. Anderson
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Juliana K. Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
7
|
Lo HY, Long DR, Holmes EA, Penewit K, Hodgson T, Lewis JD, Waalkes A, Salipante SJ. Transposon sequencing identifies genes impacting Staphylococcus aureus invasion in a human macrophage model. Infect Immun 2023; 91:e0022823. [PMID: 37676013 PMCID: PMC10580828 DOI: 10.1128/iai.00228-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 09/08/2023] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen in many host cell types, facilitating its persistence in chronic infections. The genes contributing to intracellular pathogenesis have not yet been fully enumerated. Here, we cataloged genes influencing S. aureus invasion and survival within human THP-1 derived macrophages using two laboratory strains (ATCC2913 and JE2). We developed an in vitro transposition method to produce highly saturated transposon mutant libraries in S. aureus and performed transposon insertion sequencing (Tn-Seq) to identify candidate genes with significantly altered abundance following macrophage invasion. While some significant genes were strain-specific, 108 were identified as common across both S. aureus strains, with most (n = 106) being required for optimal macrophage infection. We used CRISPR interference (CRISPRi) to functionally validate phenotypic contributions for a subset of genes. Of the 20 genes passing validation, seven had previously identified roles in S. aureus virulence, and 13 were newly implicated. Validated genes frequently evidenced strain-specific effects, yielding opposing phenotypes when knocked down in the alternative strain. Genomic analysis of de novo mutations occurring in groups (n = 237) of clonally related S. aureus isolates from the airways of chronically infected individuals with cystic fibrosis (CF) revealed significantly greater in vivo purifying selection in conditionally essential candidate genes than those not associated with macrophage invasion. This study implicates a core set of genes necessary to support macrophage invasion by S. aureus, highlights strain-specific differences in phenotypic effects of effector genes, and provides evidence for selection of candidate genes identified by Tn-Seq analyses during chronic airway infection in CF patients in vivo.
Collapse
Affiliation(s)
- Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elizbeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
8
|
Hachem AA, Filkins LM, Kidane YH, Raj P, Tareen NG, Arana CA, Muthukrishnan G, Copley LA. Staphylococcus aureus isolates from children with clinically differentiated osteomyelitis exhibit distinct transcriptomic signatures. PLoS One 2023; 18:e0288758. [PMID: 37561761 PMCID: PMC10414669 DOI: 10.1371/journal.pone.0288758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023] Open
Abstract
There is substantial genomic heterogeneity among Staphylococcus aureus isolates of children with acute hematogenous osteomyelitis (AHO) but transcriptional behavior of clinically differentiated strains has not been previously described. This study evaluates transcriptional activity of S. aureus isolates of children with AHO that may regulate metabolism, biosynthesis, or virulence during bacterial growth and pathogenesis. In vitro growth kinetics were compared between three S. aureus clinical isolates from children with AHO who had mild, moderate, and severe illness. Total RNA sequencing was performed for each isolate at six separate time points throughout the logarithmic phase of growth. The NASA RNA-Sequencing Consensus Pipeline was used to identify differentially expressed genes allowing for 54 comparisons between the three isolates during growth. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways were used to evaluate transcriptional variation in metabolism, biosynthesis pathways and virulence potential of the isolates. The S. aureus isolates demonstrated differing growth kinetics under standardized conditions with the mild isolate having higher optical densities with earlier and higher peak rates of growth than that of the other isolates (p<0.001). Enrichment pathway analysis established distinct transcriptional signatures according to both sampling time and clinical severity. Moderate and severe isolates demonstrated pathways of bacterial invasion, S. aureus infection, quorum sensing and two component systems. In comparison, the mild strain favored biosynthesis and metabolism. These findings suggest that transcriptional regulation during the growth of S. aureus may impact the pathogenetic mechanisms involved in the progression of severity of illness in childhood osteomyelitis. The clinical isolates studied demonstrated a tradeoff between growth and virulence. Further investigation is needed to evaluate these transcriptional pathways in an animal model or during active clinical infections of children with AHO.
Collapse
Affiliation(s)
- Ahmad A. Hachem
- Department of Pediatrics, University of Florida College of Medicine –Jacksonville, Jacksonville, FL, United States of America
| | - Laura M. Filkins
- Department of Microbiology, University of Texas Southwestern, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Yared H. Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, United States of America
| | - Prithvi Raj
- Microbiome Research Laboratory, University of Texas Southwestern, Dallas, TX, United States of America
| | - Naureen G. Tareen
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Carlos A. Arana
- Genomics Core, University of Texas Southwestern, Dallas, TX, United States of America
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Lawson A. Copley
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
- Department of Pediatric Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, United States of America
| |
Collapse
|
9
|
Hsieh RC, Liu R, Burgin DJ, Otto M. Understanding mechanisms of virulence in MRSA: implications for antivirulence treatment strategies. Expert Rev Anti Infect Ther 2023; 21:911-928. [PMID: 37501364 DOI: 10.1080/14787210.2023.2242585] [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: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen, often causing recurrent and deadly infections in the hospital and community. Many S. aureus virulence factors have been suggested as potential targets for antivirulence therapy to decrease the threat of diminishing antibiotic availability. Antivirulence methods hold promise due to their adjunctive and prophylactic potential and decreased risk for selective pressure. AREAS COVERED This review describes the dominant virulence mechanisms exerted by MRSA and antivirulence therapeutics that are currently undergoing testing in clinical or preclinical stages. We also discuss the advantages and downsides of several investigational antivirulence approaches, including the targeting of bacterial transporters, host-directed therapy, and quorum-sensing inhibitors. For this review, a systematic search of literature on PubMed, Google Scholar, and Web of Science for relevant search terms was performed in April and May 2023. EXPERT OPINION Vaccine and antibody strategies have failed in clinical trials and could benefit from more basic science-informed approaches. Antivirulence-targeting approaches need to be set up better to meet the requirements of drug development, rather than only providing limited results to provide 'proof-of-principle' translational value of pathogenesis research. Nevertheless, there is great potential of such strategies and potential particular promise for novel probiotic approaches.
Collapse
Affiliation(s)
- Roger C Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Dylan J Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
10
|
Buckley PT, Chan R, Fernandez J, Luo J, Lacey KA, DuMont AL, O'Malley A, Brezski RJ, Zheng S, Malia T, Whitaker B, Zwolak A, Payne A, Clark D, Sigg M, Lacy ER, Kornilova A, Kwok D, McCarthy S, Wu B, Morrow B, Nemeth-Seay J, Petley T, Wu S, Strohl WR, Lynch AS, Torres VJ. Multivalent human antibody-centyrin fusion protein to prevent and treat Staphylococcus aureus infections. Cell Host Microbe 2023; 31:751-765.e11. [PMID: 37098341 DOI: 10.1016/j.chom.2023.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 04/27/2023]
Abstract
Treating and preventing infections by antimicrobial-resistant bacterial pathogens is a worldwide problem. Pathogens such as Staphylococcus aureus produce an array of virulence determinants, making it difficult to identify single targets for the development of vaccines or monoclonal therapies. We described a human-derived anti-S. aureus monoclonal antibody (mAb)-centyrin fusion protein ("mAbtyrin") that simultaneously targets multiple bacterial adhesins, resists proteolysis by bacterial protease GluV8, avoids Fc engagement by S. aureus IgG-binding proteins SpA and Sbi, and neutralizes pore-forming leukocidins via fusion with anti-toxin centyrins, while maintaining Fc- and complement-mediated functions. Compared with the parental mAb, mAbtyrin protected human phagocytes and boosted phagocyte-mediated killing. The mAbtyrin also reduced pathology, reduced bacterial burden, and protected from different types of infections in preclinical animal models. Finally, mAbtyrin synergized with vancomycin, enhancing pathogen clearance in an animal model of bacteremia. Altogether, these data establish the potential of multivalent mAbs for treating and preventing S. aureus diseases.
Collapse
Affiliation(s)
- Peter T Buckley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA.
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Jeffrey Fernandez
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Jinquan Luo
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Ashley L DuMont
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Randall J Brezski
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Songmao Zheng
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Thomas Malia
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Brian Whitaker
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Adam Zwolak
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Angela Payne
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Desmond Clark
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Martin Sigg
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Eilyn R Lacy
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Anna Kornilova
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Debra Kwok
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Steve McCarthy
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Bingyuan Wu
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Brian Morrow
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | | | - Ted Petley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Sam Wu
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - William R Strohl
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | | | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
11
|
Grazul M, Balcerczak E, Sienkiewicz M. Analysis of the Presence of the Virulence and Regulation Genes from Staphylococcus aureus ( S. aureus) in Coagulase Negative Staphylococci and the Influence of the Staphylococcal Cross-Talk on Their Functions. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:5155. [PMID: 36982064 PMCID: PMC10049693 DOI: 10.3390/ijerph20065155] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 06/18/2023]
Abstract
Coagulase-negative staphylococci (CoNS) are increasingly becoming a public health issue worldwide due to their growing resistance to antibiotics and common involvement in complications related to invasive surgical procedures, and nosocomial and urinary tract infections. Their behavior either as a commensal or a pathogen is a result of strict regulation of colonization and virulence factors. Although functionality of virulence factors and processes involved in their regulation are quite well understood in S. aureus, little is known about them in CoNS species. Therefore, the aim of our studies was to check if clinical CoNS strains may contain virulence factors and genes involved in resistance to methicillin, that are homologous to S. aureus. Moreover, we checked the presence of elements responsible for regulation of genes that encode virulence factors typical for S. aureus in tested isolates. We also investigated whether the regulation factors produced by one CoNS isolate can affect virulence activity of other strains by co-incubation of tested isolates with supernatant from other isolates. Our studies confirmed the presence of virulence factor and regulatory genes attributed to S. aureus in CoNS isolates and indicated that one strain with an active agr gene is able to affect biofilm formation and δ-toxin activity of strains with inactive agr genes. The cognition of prevalence and regulation of virulence factors as well as antibiotic resistance of CoNS isolates is important for better control and treatment of CoNS infections.
Collapse
Affiliation(s)
- Magdalena Grazul
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| |
Collapse
|
12
|
Haag AF, Liljeroos L, Donato P, Pozzi C, Brignoli T, Bottomley MJ, Bagnoli F, Delany I. In Vivo Gene Expression Profiling of Staphylococcus aureus during Infection Informs Design of Stemless Leukocidins LukE and -D as Detoxified Vaccine Candidates. Microbiol Spectr 2023; 11:e0257422. [PMID: 36688711 PMCID: PMC9927290 DOI: 10.1128/spectrum.02574-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/02/2023] [Indexed: 01/24/2023] Open
Abstract
Staphylococcus aureus is a clinically important bacterial pathogen that has become resistant to treatment with most routinely used antibiotics. Alternative strategies, such as vaccination and phage therapy, are therefore actively being investigated to prevent or combat staphylococcal infections. Vaccination requires that vaccine targets are expressed at sufficient quantities during infection so that they can be targeted by the host's immune system. While our knowledge of in vitro expression levels of putative vaccine candidates is comprehensive, crucial in vivo expression data are scarce and promising vaccine candidates during in vitro assessment often prove ineffective in preventing S. aureus infection. Here, we show how a newly developed high-throughput quantitative reverse transcription-PCR (qRT-PCR) assay monitoring the expression of 84 staphylococcal genes encoding mostly virulence factors can inform the selection and design of effective vaccine candidates against staphylococcal infections. We show that this assay can accurately quantify mRNA expression levels of these genes in several host organs relying only on very limited amounts of bacterial mRNA in each sample. We selected two highly expressed genes, lukE and lukD, encoding pore-forming leukotoxins, to inform the design of detoxified recombinant proteins and showed that immunization with recombinant genetically detoxified LukED antigens conferred protection against staphylococcal skin infection in mice. Consequently, knowledge of in vivo-expressed virulence determinants can be successfully deployed to identify and select promising candidates for optimized design of effective vaccine antigens against S. aureus. Notably, this approach should be broadly applicable to numerous other pathogens. IMPORTANCE Vaccination is an attractive strategy for preventing bacterial infections in an age of increased antimicrobial resistance. However, vaccine development frequently suffers significant setbacks when candidate antigens that show promising results in in vitro experimentation fail to protect from disease. An alluring strategy is to focus resources on developing bacterial virulence factors that are expressed during disease establishment or maintenance and are critical for bacterial in-host survival as vaccine targets. While expression profiles of many virulence factors have been characterized in detail in vitro, our knowledge of their in vivo expression profiles is still scarce. Here, using a high-throughput qRT-PCR approach, we identified two highly expressed leukotoxins in a murine infection model and showed that genetically detoxified derivatives of these elicited a protective immune response in a murine skin infection model. Therefore, in vivo gene expression can inform the selection of promising candidates for the design of effective vaccine antigens.
Collapse
Affiliation(s)
- Andreas F. Haag
- GSK, Siena, Italy
- School of Medicine, University of St. Andrews, St. Andrews, United Kingdom
| | | | | | | | - Tarcisio Brignoli
- GSK, Siena, Italy
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | | | | |
Collapse
|
13
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
14
|
Aguilar-Gómez NE, Merida-Vieyra J, Isunza-Alonso OD, Morales-Pirela MG, Colín-Martínez O, Juárez-Benítez EJ, García de la Puente S, Aquino-Andrade A. Surveillance of osteoarticular infections caused by Staphylococcus aureus in a paediatric hospital in Mexico City. Front Cell Infect Microbiol 2022; 12:999268. [PMID: 36569208 PMCID: PMC9774039 DOI: 10.3389/fcimb.2022.999268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/20/2022] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus is the main aetiologic agent of osteoarticular infections (OAIs) in paediatric patients. The aim of this prospective unicenter study was to describe the phenotypic and genotypic characteristics of S. aureus isolates obtained from OAIs in paediatric patients admitted to tertiary care hospital. Through a surveillance program called OsteoCode, a multidisciplinary team was created and we identified 27 patients with OAIs caused by S. aureus from 2019 to 2021. The susceptibility profile, virulence factors, biofilm formation, pulsed-field gel electrophoresis (PFGE), clonal complex (CC) and sequence type (ST) were determined. In addition, the clinical characteristics and evolution of the patients presented six months after the diagnosis of OAIs were described. Ninety-two percent of the isolates were methicillin-sensitive S. aureus (MSSA). In methicillin-resistant S. aureus (MRSA), SCCmec-II and SCCmec-V were detected. The pvl gene was only observed in MSSA (18.5%) and was associated with highest fever (p=0.015), multiple localization (p=0.017), and soft tissue sites of infection beyond the bone (pyomyositis, pulmonary abscess) (p=0.017). Biofilm formation was detected in 55.6% of isolates. The most common CC were CC5 and CC30 which represent the most common linages for bone and joint infections worldwide. The isolates were distributed in different STs, and ST672 was predominant. MRSA were associated with a longer duration of intravenous treatment and a prolonged hospital stay (p=0.023). Recurrent infection occurred in five children and orthopaedic complications in 33.3% of patients. This is the first study that reflects the epidemiology of S. aureus in OAIs in paediatric patients in Mexico; a clear predominance of MSSA distributed in different STs was observed. Our findings highlight that a multidisciplinary team is required for the diagnosis and treatment of OAIs.
Collapse
Affiliation(s)
| | - Jocelin Merida-Vieyra
- Molecular Microbiology Laboratory, Instituto Nacional de Pediatria, Mexico City, Mexico
| | | | | | - Oscar Colín-Martínez
- Department of Orthopaedic Surgery, Instituto Nacional de Pediatria, Mexico City, Mexico
| | | | | | - Alejandra Aquino-Andrade
- Molecular Microbiology Laboratory, Instituto Nacional de Pediatria, Mexico City, Mexico,*Correspondence: Alejandra Aquino-Andrade,
| |
Collapse
|
15
|
Staphylococcus lugdunensis Uses the Agr Regulatory System to Resist Killing by Host Innate Immune Effectors. Infect Immun 2022; 90:e0009922. [PMID: 36069592 PMCID: PMC9584346 DOI: 10.1128/iai.00099-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Coagulase-negative staphylococci (CoNS) are frequently commensal bacteria that rarely cause disease in mammals. Staphylococcus lugdunensis is an exceptional CoNS that causes disease in humans similar to virulent Staphylococcus aureus, but the factors that enhance the virulence of this bacterium remain ill defined. Here, we used random transposon insertion mutagenesis to identify the agr quorum sensing system as a regulator of hemolysins in S. lugdunensis. Using RNA sequencing (RNA-seq), we revealed that agr regulates dozens of genes, including hemolytic S. lugdunensis synergistic hemolysins (SLUSH) peptides and the protease lugdulysin. A murine bacteremia model was used to show that mice infected systemically with wild-type S. lugdunensis do not show overt signs of disease despite there being high numbers of bacteria in the livers and kidneys of mice. Moreover, proliferation of the agr mutant in these organs was no different from that of the wild-type strain, leaving the role of the SLUSH peptides and the metalloprotease lugdulysin in pathogenesis still unclear. Nonetheless, the tropism of S. lugdunensis for humans led us to investigate the role of virulence factors in other ways. We show that agr-regulated effectors, but not SLUSH or lugdulysin alone, are important for S. lugdunensis survival in whole human blood. Moreover, we demonstrate that Agr contributes to survival of S. lugdunensis during encounters with murine and primary human macrophages. These findings demonstrate that, in S. lugdunensis, Agr regulates expression of virulence factors and is required for resistance to host innate antimicrobial defenses. This study therefore provides insight into strategies that this Staphylococcus species uses to cause disease.
Collapse
|
16
|
Zwack EE, Chen Z, Devlin JC, Li Z, Zheng X, Weinstock A, Lacey KA, Fisher EA, Fenyö D, Ruggles KV, Loke P, Torres VJ. Staphylococcus aureus induces a muted host response in human blood that blunts the recruitment of neutrophils. Proc Natl Acad Sci U S A 2022; 119:e2123017119. [PMID: 35881802 PMCID: PMC9351360 DOI: 10.1073/pnas.2123017119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/29/2022] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen and chief among bloodstream-infecting bacteria. S. aureus produces an array of human-specific virulence factors that may contribute to immune suppression. Here, we defined the response of primary human phagocytes following infection with S. aureus using RNA-sequencing (RNA-Seq). We found that the overall transcriptional response to S. aureus was weak both in the number of genes and in the magnitude of response. Using an ex vivo bacteremia model with fresh human blood, we uncovered that infection with S. aureus resulted in the down-regulation of genes related to innate immune response and cytokine and chemokine signaling. This muted transcriptional response was conserved across diverse S. aureus clones but absent in blood exposed to heat-killed S. aureus or blood infected with the less virulent staphylococcal species Staphylococcus epidermidis. Notably, this signature was also present in patients with S. aureus bacteremia. We identified the master regulator S. aureus exoprotein expression (SaeRS) and the SaeRS-regulated pore-forming toxins as key mediators of the transcriptional suppression. The S. aureus-mediated suppression of chemokine and cytokine transcription was reflected by circulating protein levels in the plasma. Wild-type S. aureus elicited a soluble milieu that was restrictive in the recruitment of human neutrophils compared with strains lacking saeRS. Thus, S. aureus blunts the inflammatory response resulting in impaired neutrophil recruitment, which could promote the survival of the pathogen during invasive infection.
Collapse
Affiliation(s)
- Erin E. Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Ze Chen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Joseph C. Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Zhi Li
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016
| | - Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Ada Weinstock
- Department of Medicine Cardiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Edward A. Fisher
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
- Department of Medicine Cardiology, New York University Grossman School of Medicine, New York, NY 10016
| | - David Fenyö
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016
- Department for Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016
| | - Kelly V. Ruggles
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016
- Division of Translational Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016
| | - P’ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
17
|
Muneeb KH, Sudha S, Sivaraman GK, Ojha R, Mendem SK, Murugesan D, Raisen CL, Shome B, Holmes M. Whole-genome sequence analysis of Staphylococcus aureus from retail fish acknowledged the incidence of highly virulent ST672-MRSA-IVa/t1309, an emerging Indian clone, in Assam, India. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:412-421. [PMID: 34796671 DOI: 10.1111/1758-2229.13024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 11/04/2021] [Indexed: 06/13/2023]
Abstract
The epidemiology and toxigenicity of MRSA in the fishery environment are poorly understood. In this study, methicillin-resistant Staphylococcus aureus (MRSA) (n = 1) and methicillin-susceptible S. aureus (MSSA) (n = 2) from retail fish were subjected to comprehensive genome analysis. Here, we report the occurrence of ST672-MRSA-IV/t1309 and ST5-MSSA/t105 for the first time from India in the fishery environment. The resistome of the isolates was in concordance with their phenotypic resistance pattern. Phenotypically, the resistance profile of MSSA isolates (n = 2) was AMP-CLI-ERY-NOR-PEN. For MRSA (n = 1), it was AMP-CFZ-CLI-ERY-NOR-OXA-PEN. The antibiotic efflux genes and mutations in the antibiotic target accounted for fluoroquinolone resistance whereas methicillin resistance was conferred through possession of a mecA gene. Similarly, all three isolates carried a similar array of virulence factors. The conjugative plasmid inc18 and rep family 10 plasmids were found in two of the three isolates. This study documents the MRSA carrying SCCmec IVa elements which are the markers of community-associated MRSA (CA-MRSA). Through the possession of SCCmec IV elements, which are smaller than other types of SCCmec, MRSA can contribute to the rapid dissemination of antimicrobial resistance and virulence factors. In short, our findings highlighted that the presence of ST672-MRSA in fishery environments may pose a risk to human health.
Collapse
Affiliation(s)
- K H Muneeb
- Microbiology Fermentation and Biotechnology Division, ICAR-Central Institute of Fisheries Technology, Kochi, 682 029, India
- Department of Biotechnology, Faculty of Sciences, Cochin University of Science and Technology, Kochi, India
| | - S Sudha
- Microbiology Fermentation and Biotechnology Division, ICAR-Central Institute of Fisheries Technology, Kochi, 682 029, India
| | - G K Sivaraman
- Microbiology Fermentation and Biotechnology Division, ICAR-Central Institute of Fisheries Technology, Kochi, 682 029, India
| | - Rakshit Ojha
- Department of Disease Investigation, ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bangalore, India
| | - Suresh Kumar Mendem
- Department of Disease Investigation, ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bangalore, India
| | - Devi Murugesan
- Department of Disease Investigation, ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bangalore, India
| | - C L Raisen
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Bibek Shome
- Department of Disease Investigation, ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bangalore, India
| | - Mark Holmes
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most common causes of hospital-acquired pneumonia. To better manage patients with MRSA pneumonia, we require a greater understanding of the host-pathogen interactions during infection. MRSA research focuses on highly virulent and cytotoxic strains, which demonstrate robust phenotypes in animal models of infection. However, nosocomial infections are often caused by hospital-acquired MRSA (HA-MRSA) isolates that exhibit low cytotoxicity and few or no phenotypes in mice, thereby confounding mechanistic studies of pathogenesis. Consequently, virulence pathways utilized by HA-MRSA in nosocomial pneumonia are largely unknown. Here, we report that conditioning mice with broad-spectrum antibiotics lowers the barrier to pneumonia, thereby transforming otherwise avirulent HA-MRSA isolates into lethal pathogens. HA-MRSA isolates are avirulent in gnotobiotic mice, mimicking results in conventional animals. Thus, the observed enhanced susceptibility to infection in antibiotic-treated mice is not due to depletion of the microbiota. More generally, we found that antibiotic conditioning leads to increased susceptibility to infection by diverse antimicrobial-resistant (AMR) pathogens of low virulence. Treatment with antibiotics leads to dehydration and malnutrition, suggesting a potential role for these clinically relevant and reducible hospital complications in susceptibility to pathogens. In sum, the model described here mitigates the impact of low virulence in immunocompetent mice, providing a convenient model to gain fundamental insight into the pathogenesis of nosocomial pathogens.
Collapse
|
19
|
Zheng X, Ma SX, St. John A, Torres VJ. The Major Autolysin Atl Regulates the Virulence of Staphylococcus aureus by Controlling the Sorting of LukAB. Infect Immun 2022; 90:e0005622. [PMID: 35258336 PMCID: PMC9022505 DOI: 10.1128/iai.00056-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/14/2023] Open
Abstract
Infections caused by the Gram-positive bacterium Staphylococcus aureus remain a significant health threat globally. The production of bicomponent pore-forming leukocidins plays an important role in S. aureus pathogenesis. Transcriptionally, these toxins are primarily regulated by the Sae and Agr regulatory systems. However, the posttranslational regulation of these toxins is largely unexplored. In particular, one of the leukocidins, LukAB, has been shown to be both secreted into the extracellular milieu and associated with the bacterial cell envelope. Here, we report that a major cell wall hydrolase, autolysin (Atl), controls the sorting of LukAB from the cell envelope to the extracellular milieu, an effect independent of transcriptional regulation. By influencing the sorting of LukAB, Atl modulates S. aureus cytotoxicity toward primary human neutrophils. Mechanistically, we found that the reduction in peptidoglycan cleavage and increased LukAB secretion in the atl mutant can be reversed through the supplementation of exogenous mutanolysin. Altogether, our study revealed that the cell wall hydrolase activity of Atl and the cleavage of peptidoglycan play an important role in controlling the sorting of S. aureus toxins during secretion.
Collapse
Affiliation(s)
- Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sheya Xiao Ma
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Amelia St. John
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Boulouis C, Leeansyah E, Mairpady Shambat S, Norrby-Teglund A, Sandberg JK. Mucosa-Associated Invariant T Cell Hypersensitivity to Staphylococcus aureus Leukocidin ED and Its Modulation by Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1170-1179. [PMID: 35140134 PMCID: PMC9012079 DOI: 10.4049/jimmunol.2100912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
Mucosa-associated invariant T (MAIT) cells recognize bacterial riboflavin metabolite Ags presented by MHC class Ib-related protein (MR1) and play important roles in immune control of microbes that synthesize riboflavin. This includes the pathobiont Staphylococcus aureus, which can also express a range of virulence factors, including the secreted toxin leukocidin ED (LukED). In this study, we found that human MAIT cells are hypersensitive to LukED-mediated lysis and lost on exposure to the toxin, leaving a T cell population devoid of MAIT cells. The cytolytic effect of LukED on MAIT cells was rapid and occurred at toxin concentrations lower than those required for toxicity against conventional T cells. Furthermore, this coincided with high MAIT cell expression of CCR5, and loss of these cells was efficiently inhibited by the CCR5 inhibitor maraviroc. Interestingly, exposure and preactivation of MAIT cells with IL-12 and IL-18, or activation via TCR triggering, partially protected from LukED toxicity. Furthermore, analysis of NK cells indicated that LukED targeted the mature cytotoxic CD57+ NK cell subset in a CCR5-independent manner. Overall, these results indicate that LukED efficiently eliminates immune cells that can respond rapidly to S. aureus in an innate fashion without the need for clonal expansion, and that MAIT cells are exceptionally vulnerable to this toxin. Thus, the findings support a model where LukED secretion may allow S. aureus to avoid recognition by the rapid cell-mediated responses mediated by MAIT cells and NK cells.
Collapse
Affiliation(s)
- Caroline Boulouis
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Edwin Leeansyah
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, People's Republic of China; and
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, People's Republic of China
| | | | - Anna Norrby-Teglund
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden;
| |
Collapse
|
21
|
Ul-Hamid A, Dafalla H, Hakeem AS, Haider A, Ikram M. In-Vitro Catalytic and Antibacterial Potential of Green Synthesized CuO Nanoparticles against Prevalent Multiple Drug Resistant Bovine Mastitogen Staphylococcus aureus. Int J Mol Sci 2022; 23:2335. [PMID: 35216450 PMCID: PMC8878101 DOI: 10.3390/ijms23042335] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 01/07/2023] Open
Abstract
Nanoparticles prepared from bio-reduction agents are of keen interest to researchers around the globe due to their ability to mitigate the harmful effects of chemicals. In this regard, the present study aims to synthesize copper oxide nanoparticles (CuO NPs) by utilizing root extracts of ginger and garlic as reducing agents, followed by the characterization and evaluation of their antimicrobial properties against multiple drug resistant (MDR) S. aureus. In this study, UV-vis spectroscopy revealed a reduced degree of absorption with an increase in the extract amount present in CuO. The maximum absorbance for doped NPs was recorded around 250 nm accompanying redshift. X-ray diffraction analysis revealed the monoclinic crystal phase of the particles. The fabricated NPs exhibited spherical shapes with dense agglomeration when examined with FE-SEM and TEM. The crystallite size measured by using XRD was found to be within a range of 23.38-46.64 nm for ginger-doped CuO and 26-56 nm for garlic-doped CuO. Green synthesized NPs of ginger demonstrated higher bactericidal tendencies against MDR S. aureus. At minimum and maximum concentrations of ginger-doped CuO NPs, substantial inhibition areas for MDR S. aureus were (2.05-3.80 mm) and (3.15-5.65 mm), and they were measured as (1.1-3.55 mm) and (1.25-4.45 mm) for garlic-doped NPs. Conventionally available CuO and crude aqueous extract (CAE) of ginger and garlic roots reduced MB in 12, 21, and 38 min, respectively, in comparison with an efficient (100%) reduction of dye in 1 min and 15 s for ginger and garlic doped CuO NPs.
Collapse
Affiliation(s)
- Anwar Ul-Hamid
- Core Research Facilities, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia;
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| | - Hatim Dafalla
- Core Research Facilities, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia;
| | - Abbas Saeed Hakeem
- Interdisciplinary Research Center for Hydrogen and Energy Storage, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia;
| | - Ali Haider
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture (MNSUA), Multan 66000, Pakistan;
| | - Muhammad Ikram
- Solar Cell Applications Research Lab, Department of Physics, Government College University, Lahore 54000, Pakistan;
| |
Collapse
|
22
|
Single-Chain Fragment Variables Targeting Leukocidin ED Can Alleviate the Inflammation of Staphylococcus aureus-Induced Mastitis in Mice. Int J Mol Sci 2021; 23:ijms23010334. [PMID: 35008761 PMCID: PMC8745144 DOI: 10.3390/ijms23010334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus aureus is a vital bovine mastitis pathogen causing huge economic losses to the dairy industry worldwide. In our previous studies, leukotoxin ED (LukED) was detected in most S. aureus strains isolated from bovine mastitis. Here, four single-chain fragment variables (scFvs) (ZL8 and ZL42 targeting LukE, ZL22 and ZL23 targeting LukD) were obtained using purified LukE and LukD proteins as the antigens after five rounds of bio-panning. The complementarity-determining region 3 (CDR3) of the VH domain of these scFvs exhibited significant diversities. In vitro, the scFvs significantly decreased LukED-induced cell killing by inhibiting the binding of LukED to chemokine receptors (CCR5 and CXCR2) and reduced the death rates of bovine neutrophils and MAC-T cells caused by LukED and S. aureus (p < 0.05). In an S. aureus-induced mouse mastitis model, histopathology and MPO results revealed that scFvs ameliorated the histopathological damages and reduced the infiltration of inflammatory cells (p < 0.05). The ELISA and qPCR assays showed that scFvs reduced the transcription and expression levels of Tumor Necrosis Factor-alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-8 and IL-18 (p < 0.05). The overall results demonstrated the protective anti-inflammatory effect of scFvs in vitro and in vivo, enlightening the potential role of scFvs in the prevention and treatment of S. aureus-induced mastitis.
Collapse
|
23
|
Anti- Staphylococcus aureus Single-Chain Fragment Variables Play a Protective Anti-Inflammatory Role In Vitro and In Vivo. Vaccines (Basel) 2021; 9:vaccines9111300. [PMID: 34835231 PMCID: PMC8618225 DOI: 10.3390/vaccines9111300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is a causative agent of bovine mastitis, capable of causing significant economic losses to the dairy industry worldwide. This study focuses on obtaining single-chain fragment variables (scFvs) against the virulence factors of S. aureus and evaluates the protective effect of scFvs on bovine mammary epithelial (MAC-T) cells and mice mammary gland tissues infected by S. aureus. After five rounds of bio-panning, four scFvs targeting four virulence factors of S. aureus were obtained. The complementarity-determining regions (CDRs) of these scFvs exhibited significant diversities, especially CDR3 of the VH domain. In vitro, each of scFvs was capable of inhibiting S. aureus growth and reducing the damage of MAC-T cells infected by S. aureus. Preincubation of MAC-T cells with scFvs could significantly attenuate the effect of apoptosis and necrosis compared with the negative control group. In vivo, the qPCR and ELISA results demonstrated that scFvs reduced the transcription and expression of Tumor Necrosis Factor alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-8, and IL-18. Histopathology and myeloperoxidase (MPO) results showed that scFvs ameliorated the histopathological damages and reduced the inflammatory cells infiltration. The overall results demonstrated the positive anti-inflammatory effect of scFvs, revealing the potential role of scFvs in the prevention and treatment of S. aureus infections.
Collapse
|
24
|
The cell envelope of Staphylococcus aureus selectively controls the sorting of virulence factors. Nat Commun 2021; 12:6193. [PMID: 34702812 PMCID: PMC8548510 DOI: 10.1038/s41467-021-26517-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.
Collapse
|
25
|
Cruz AR, van Strijp JAG, Bagnoli F, Manetti AGO. Virulence Gene Expression of Staphylococcus aureus in Human Skin. Front Microbiol 2021; 12:692023. [PMID: 34177874 PMCID: PMC8231915 DOI: 10.3389/fmicb.2021.692023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/19/2021] [Indexed: 01/03/2023] Open
Abstract
Staphylococcus aureus is the main cause of human skin and soft tissue infections. However, S. aureus pathogenicity within the skin is not fully characterized. Here, we implemented an S. aureus cutaneous infection model using human skin explants and performed a time-course infection to study the gene expression profile of a large panel of virulence-related factors of S. aureus USA300 LAC strain, by high-throughput RT-PCR. We pinpointed the genes that were differentially regulated by the bacteria in the skin tissues and identified 12 virulence factors that were upregulated at all time points assessed. Finally, using confocal microscopy, we show that the expression of alpha-hemolysin by S. aureus varies dependent on the skin niche and that the bacteria preferentially accumulates inside sweat glands and ducts. Taken together, our study gives insights about the pathogenic lifestyle of S. aureus within human skin tissues, which may contribute for the development of anti-S. aureus therapeutic strategies.
Collapse
Affiliation(s)
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | |
Collapse
|
26
|
Perelman SS, James DBA, Boguslawski KM, Nelson CW, Ilmain JK, Zwack EE, Prescott RA, Mohamed A, Tam K, Chan R, Narechania A, Pawline MB, Vozhilla N, Moustafa AM, Kim SY, Dittmann M, Ekiert DC, Bhabha G, Shopsin B, Planet PJ, Koralov SB, Torres VJ. Genetic variation of staphylococcal LukAB toxin determines receptor tropism. Nat Microbiol 2021; 6:731-745. [PMID: 33875847 PMCID: PMC8597016 DOI: 10.1038/s41564-021-00890-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 03/11/2021] [Indexed: 02/02/2023]
Abstract
Staphylococcus aureus has evolved into diverse lineages, known as clonal complexes (CCs), which exhibit differences in the coding sequences of core virulence factors. Whether these alterations affect functionality is poorly understood. Here, we studied the highly polymorphic pore-forming toxin LukAB. We discovered that the LukAB toxin variants produced by S. aureus CC30 and CC45 kill human phagocytes regardless of whether CD11b, the previously established LukAB receptor, is present, and instead target the human hydrogen voltage-gated channel 1 (HVCN1). Biochemical studies identified the domain within human HVCN1 that drives LukAB species specificity, enabling the generation of humanized HVCN1 mice with enhanced susceptibility to CC30 LukAB and to bloodstream infection caused by CC30 S. aureus strains. Together, this work advances our understanding of an important S. aureus toxin and underscores the importance of considering genetic variation in characterizing virulence factors and understanding the tug of war between pathogens and the host.
Collapse
Affiliation(s)
- Sofya S Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - David B A James
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Chase W Nelson
- Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Juliana K Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Erin E Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rachel A Prescott
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Adil Mohamed
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kayan Tam
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Apurva Narechania
- Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
| | - Miranda B Pawline
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY, USA
| | - Nikollaq Vozhilla
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ahmed M Moustafa
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sang Y Kim
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Office of Collaborative Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Meike Dittmann
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Damian C Ekiert
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Gira Bhabha
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY, USA
| | - Paul J Planet
- Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sergei B Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Klebba PE, Newton SMC, Six DA, Kumar A, Yang T, Nairn BL, Munger C, Chakravorty S. Iron Acquisition Systems of Gram-negative Bacterial Pathogens Define TonB-Dependent Pathways to Novel Antibiotics. Chem Rev 2021; 121:5193-5239. [PMID: 33724814 PMCID: PMC8687107 DOI: 10.1021/acs.chemrev.0c01005] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Iron is an indispensable metabolic cofactor in both pro- and eukaryotes, which engenders a natural competition for the metal between bacterial pathogens and their human or animal hosts. Bacteria secrete siderophores that extract Fe3+ from tissues, fluids, cells, and proteins; the ligand gated porins of the Gram-negative bacterial outer membrane actively acquire the resulting ferric siderophores, as well as other iron-containing molecules like heme. Conversely, eukaryotic hosts combat bacterial iron scavenging by sequestering Fe3+ in binding proteins and ferritin. The variety of iron uptake systems in Gram-negative bacterial pathogens illustrates a range of chemical and biochemical mechanisms that facilitate microbial pathogenesis. This document attempts to summarize and understand these processes, to guide discovery of immunological or chemical interventions that may thwart infectious disease.
Collapse
Affiliation(s)
- Phillip E Klebba
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Salete M C Newton
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - David A Six
- Venatorx Pharmaceuticals, Inc., 30 Spring Mill Drive, Malvern, Pennsylvania 19355, United States
| | - Ashish Kumar
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Taihao Yang
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Brittany L Nairn
- Department of Biological Sciences, Bethel University, 3900 Bethel Drive, St. Paul, Minnesota 55112, United States
| | - Colton Munger
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Somnath Chakravorty
- Jacobs School of Medicine and Biomedical Sciences, SUNY Buffalo, Buffalo, New York 14203, United States
| |
Collapse
|
28
|
Banerji R, Karkee A, Kanojiya P, Saroj SD. Pore-forming toxins of foodborne pathogens. Compr Rev Food Sci Food Saf 2021; 20:2265-2285. [PMID: 33773026 DOI: 10.1111/1541-4337.12737] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 01/04/2023]
Abstract
Pore-forming toxins (PFTs) are water-soluble molecules that have been identified as the most crucial virulence factors during bacterial pathogenesis. PFTs disrupt the host cell membrane to internalize or to deliver other bacterial or virulence factors for establishing infections. Disruption of the host cell membrane by PFTs can lead to uncontrollable exchanges between the extracellular and the intracellular matrix, thereby disturbing the cellular homeostasis. Recent studies have provided insights into the molecular mechanism of PFTs during pathogenesis. Evidence also suggests the activation of several signal transduction pathways in the host cell on recognition of PFTs. Additionally, numerous distinctive host defense mechanisms as well as membrane repair mechanisms have been reported; however, studies reveal that PFTs aid in host immune evasion of the bacteria through numerous pathways. PFTs have been primarily associated with foodborne pathogens. Infection and death from diseases by consuming contaminated food are a constant threat to public health worldwide, affecting socioeconomic development. Moreover, the emergence of new foodborne pathogens has led to the rise of bacterial antimicrobial resistance affecting the population. Hence, this review focuses on the role of PFTs secreted by foodborne pathogens. The review highlights the molecular mechanism of foodborne bacterial PFTs, assisting bacterial survival from the host immune responses and understanding the downstream mechanism in the activation of various signaling pathways in the host upon PFT recognition. PFT research is a remarkable and an important field for exploring novel and broad applications of antimicrobial compounds as therapeutics.
Collapse
Affiliation(s)
- Rajashri Banerji
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Astha Karkee
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Poonam Kanojiya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
29
|
Tam K, Lacey KA, Devlin JC, Coffre M, Sommerfield A, Chan R, O'Malley A, Koralov SB, Loke P, Torres VJ. Targeting leukocidin-mediated immune evasion protects mice from Staphylococcus aureus bacteremia. J Exp Med 2021; 217:151907. [PMID: 32602902 PMCID: PMC7478724 DOI: 10.1084/jem.20190541] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/05/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is responsible for various diseases in humans, and recurrent infections are commonly observed. S. aureus produces an array of bicomponent pore-forming toxins that target and kill leukocytes, known collectively as the leukocidins. The contribution of these leukocidins to impair the development of anti–S. aureus adaptive immunity and facilitate reinfection is unclear. Using a murine model of recurrent bacteremia, we demonstrate that infection with a leukocidin mutant results in increased levels of anti–S. aureus antibodies compared with mice infected with the WT parental strain, indicating that leukocidins negatively impact the generation of anti–S. aureus antibodies in vivo. We hypothesized that neutralizing leukocidin-mediated immune subversion by vaccination may shift this host-pathogen interaction in favor of the host. Leukocidin-immunized mice produce potent leukocidin-neutralizing antibodies and robust Th1 and Th17 responses, which collectively protect against bloodstream infections. Altogether, these results demonstrate that blocking leukocidin-mediated immune evasion can promote host protection against S. aureus bloodstream infection.
Collapse
Affiliation(s)
- Kayan Tam
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Joseph C Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Maryaline Coffre
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Alexis Sommerfield
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Aidan O'Malley
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Sergei B Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - P'ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY.,Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
30
|
Cheung GYC, Bae JS, Liu R, Hunt RL, Zheng Y, Otto M. Bacterial virulence plays a crucial role in MRSA sepsis. PLoS Pathog 2021; 17:e1009369. [PMID: 33630954 PMCID: PMC7942999 DOI: 10.1371/journal.ppat.1009369] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 03/09/2021] [Accepted: 02/10/2021] [Indexed: 12/29/2022] Open
Abstract
Bacterial sepsis is a major global cause of death. However, the pathophysiology of sepsis has remained poorly understood. In industrialized nations, Staphylococcus aureus represents the pathogen most commonly associated with mortality due to sepsis. Because of the alarming spread of antibiotic resistance, anti-virulence strategies are often proposed to treat staphylococcal sepsis. However, we do not yet completely understand if and how bacterial virulence contributes to sepsis, which is vital for a thorough assessment of such strategies. We here examined the role of virulence and quorum-sensing regulation in mouse and rabbit models of sepsis caused by methicillin-resistant S. aureus (MRSA). We determined that leukopenia was a predictor of disease outcome during an early critical stage of sepsis. Furthermore, in device-associated infection as the most frequent type of staphylococcal blood infection, quorum-sensing deficiency resulted in significantly higher mortality. Our findings give important guidance regarding anti-virulence drug development strategies for the treatment of staphylococcal sepsis. Moreover, they considerably add to our understanding of how bacterial sepsis develops by revealing a critical early stage of infection during which the battle between bacteria and leukocytes determines sepsis outcome. While sepsis has traditionally been attributed mainly to host factors, our study highlights a key role of the invading pathogen and its virulence mechanisms.
Collapse
Affiliation(s)
- Gordon Y. C. Cheung
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Justin S. Bae
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rachelle L. Hunt
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yue Zheng
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
31
|
Manyahi J, Moyo SJ, Aboud S, Langeland N, Blomberg B. Predominance of PVL-negative community-associated methicillin-resistant Staphylococcus aureus sequence type 8 in newly diagnosed HIV-infected adults, Tanzania. Eur J Clin Microbiol Infect Dis 2021; 40:1477-1485. [PMID: 33586013 PMCID: PMC8206053 DOI: 10.1007/s10096-021-04160-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/11/2021] [Indexed: 11/24/2022]
Abstract
Difficult-to-treat infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are of concern in people living with HIV infection as they are more vulnerable to infection. We aimed to identify molecular characteristics of MRSA colonizing newly diagnosed HIV-infected adults in Tanzania. Individuals newly diagnosed with HIV infection were recruited in Dar es Salaam, Tanzania, from April 2017 to May 2018, as part of the randomized clinical trial CoTrimResist (ClinicalTrials.gov identifier: NCT03087890). Nasal/nasopharyngeal isolates of Staphylococcus aureus were susceptibility tested by disk diffusion method, and cefoxitin-resistant isolates were characterized by short-reads whole genome sequencing. Four percent (22/537) of patients carried MRSA in the nose/nasopharynx. MRSA isolates were frequently resistant towards gentamicin (95%), ciprofloxacin (91%), and erythromycin (82%) but less often towards trimethoprim-sulfamethoxazole (9%). Seventy-three percent had inducible clindamycin resistance. Erythromycin-resistant isolates harbored ermC (15/18) and LmrS (3/18) resistance genes. Ciprofloxacin resistance was mediated by mutations of the quinolone resistance-determining region (QRDR) sequence in the gyrA (S84L) and parC (S80Y) genes. All isolates belonged to the CC8 and ST8-SCCmecIV MRSA clone. Ninety-five percent of the MRSA isolates were spa-type t1476, and one exhibited spa-type t064. All isolates were negative for Panton-Valentine leucocidin (PVL) and arginine catabolic mobile element (ACME) type 1. All ST8-SCCmecIV-spa-t1476 MRSA clones from Tanzania were unrelated to the globally successful USA300 clone. Carriage of ST8 MRSA (non-USA300) was common among newly diagnosed HIV-infected adults in Tanzania. Frequent co-resistance to non-beta lactam antibiotics limits therapeutic options when infection occurs.
Collapse
Affiliation(s)
- Joel Manyahi
- Department of Clinical Science, University of Bergen, Bergen, Norway. .,National Advisory Unit for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway. .,Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, MUHAS, P.O. Box 65001, Dar es Salaam, Tanzania.
| | - Sabrina J Moyo
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, MUHAS, P.O. Box 65001, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, MUHAS, P.O. Box 65001, Dar es Salaam, Tanzania
| | - Nina Langeland
- Department of Clinical Science, University of Bergen, Bergen, Norway.,National Advisory Unit for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bjørn Blomberg
- Department of Clinical Science, University of Bergen, Bergen, Norway.,National Advisory Unit for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
32
|
Therapeutic Antibodies for the Treatment of Respiratory Tract Infections-Current Overview and Perspectives. Vaccines (Basel) 2021; 9:vaccines9020151. [PMID: 33668613 PMCID: PMC7917879 DOI: 10.3390/vaccines9020151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Respiratorytract infections (RTIs) are frequent and life-threatening diseases, accounting for several millions of deaths worldwide. RTIs implicate microorganisms, including viruses (influenza virus, coronavirus, respiratory syncytial virus (RSV)), bacteria (Pseudomonas aeruginosa, Streptococcus pneumoniae, Staphylococcus aureus and Bacillus anthracis) and fungi (Pneumocystis spp., Aspergillus spp. and very occasionally Candida spp.). The emergence of new pathogens, like the coronavirus SARS-CoV-2, and the substantial increase in drug resistance have highlighted the critical necessity to develop novel anti-infective molecules. In this context, antibodies (Abs) are becoming increasingly important in respiratory medicine and may fulfill the unmet medical needs of RTIs. However, development of Abs for treating infectious diseases is less advanced than for cancer and inflammatory diseases. Currently, only three Abs have been marketed for RTIs, namely, against pulmonary anthrax and RSV infection, while several clinical and preclinical studies are in progress. This article gives an overview of the advances in the use of Abs for the treatment of RTIs, based on the analysis of clinical studies in this field. It describes the Ab structure, function and pharmacokinetics, and discusses the opportunities offered by the various Ab formats, Ab engineering and co-treatment strategies. Including the most recent literature, it finally highlights the strengths, weaknesses and likely future trends of a novel anti-RTI Ab armamentarium.
Collapse
|
33
|
Interplay between ESKAPE Pathogens and Immunity in Skin Infections: An Overview of the Major Determinants of Virulence and Antibiotic Resistance. Pathogens 2021; 10:pathogens10020148. [PMID: 33540588 PMCID: PMC7912840 DOI: 10.3390/pathogens10020148] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
The skin is the largest organ in the human body, acting as a physical and immunological barrier against pathogenic microorganisms. The cutaneous lesions constitute a gateway for microbial contamination that can lead to chronic wounds and other invasive infections. Chronic wounds are considered as serious public health problems due the related social, psychological and economic consequences. The group of bacteria known as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter sp.) are among the most prevalent bacteria in cutaneous infections. These pathogens have a high level of incidence in hospital environments and several strains present phenotypes of multidrug resistance. In this review, we discuss some important aspects of skin immunology and the involvement of ESKAPE in wound infections. First, we introduce some fundamental aspects of skin physiology and immunology related to cutaneous infections. Following this, the major virulence factors involved in colonization and tissue damage are highlighted, as well as the most frequently detected antimicrobial resistance genes. ESKAPE pathogens express several virulence determinants that overcome the skin's physical and immunological barriers, enabling them to cause severe wound infections. The high ability these bacteria to acquire resistance is alarming, particularly in the hospital settings where immunocompromised individuals are exposed to these pathogens. Knowledge about the virulence and resistance markers of these species is important in order to develop new strategies to detect and treat their associated infections.
Collapse
|
34
|
Vasquez MT, Lubkin A, Reyes-Robles T, Day CJ, Lacey KA, Jennings MP, Torres VJ. Identification of a domain critical for Staphylococcus aureus LukED receptor targeting and lysis of erythrocytes. J Biol Chem 2020; 295:17241-17250. [PMID: 33051210 PMCID: PMC7863875 DOI: 10.1074/jbc.ra120.015757] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Leukocidin ED (LukED) is a pore-forming toxin produced by Staphylococcus aureus, which lyses host cells and promotes virulence of the bacteria. LukED enables S. aureus to acquire iron by lysing erythrocytes, which depends on targeting the host receptor Duffy antigen receptor for chemokines (DARC). The toxin also targets DARC on the endothelium, contributing to the lethality observed during bloodstream infection in mice. LukED is comprised of two monomers: LukE and LukD. LukE binds to DARC and facilitates hemolysis, but the closely related Panton-Valentine leukocidin S (LukS-PV) does not bind to DARC and is not hemolytic. The interaction of LukE with DARC and the role this plays in hemolysis are incompletely characterized. To determine the domain(s) of LukE that are critical for DARC binding, we studied the hemolytic function of LukE-LukS-PV chimeras, in which areas of sequence divergence (divergence regions, or DRs) were swapped between the toxins. We found that two regions of LukE's rim domain contribute to hemolysis, namely residues 57-75 (DR1) and residues 182-196 (DR4). Interestingly, LukE DR1 is sufficient to render LukS-PV capable of DARC binding and hemolysis. Further, LukE, by binding DARC through DR1, promotes the recruitment of LukD to erythrocytes, likely by facilitating LukED oligomer formation. Finally, we show that LukE targets murine Darc through DR1 in vivo to cause host lethality. These findings expand our biochemical understanding of the LukE-DARC interaction and the role that this toxin-receptor pair plays in S. aureus pathophysiology.
Collapse
Affiliation(s)
- Marilyn T Vasquez
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ashira Lubkin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Tamara Reyes-Robles
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Christopher J Day
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Michael P Jennings
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
35
|
Vlaeminck J, Raafat D, Surmann K, Timbermont L, Normann N, Sellman B, van Wamel WJB, Malhotra-Kumar S. Exploring Virulence Factors and Alternative Therapies against Staphylococcus aureus Pneumonia. Toxins (Basel) 2020; 12:toxins12110721. [PMID: 33218049 PMCID: PMC7698915 DOI: 10.3390/toxins12110721] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/12/2020] [Accepted: 11/15/2020] [Indexed: 12/13/2022] Open
Abstract
Pneumonia is an acute pulmonary infection associated with high mortality and an immense financial burden on healthcare systems. Staphylococcus aureus is an opportunistic pathogen capable of inducing S. aureus pneumonia (SAP), with some lineages also showing multidrug resistance. Given the high level of antibiotic resistance, much research has been focused on targeting S. aureus virulence factors, including toxins and biofilm-associated proteins, in an attempt to develop effective SAP therapeutics. Despite several promising leads, many hurdles still remain for S. aureus vaccine research. Here, we review the state-of-the-art SAP therapeutics, highlight their pitfalls, and discuss alternative approaches of potential significance and future perspectives.
Collapse
Affiliation(s)
- Jelle Vlaeminck
- Laboratory of Medical Microbiology, Vaccine and Infectious Diseases Institute, University of Antwerp, 2610 Antwerp, Belgium; (J.V.); (L.T.)
| | - Dina Raafat
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany; (D.R.); (N.N.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Kristin Surmann
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Leen Timbermont
- Laboratory of Medical Microbiology, Vaccine and Infectious Diseases Institute, University of Antwerp, 2610 Antwerp, Belgium; (J.V.); (L.T.)
| | - Nicole Normann
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany; (D.R.); (N.N.)
| | - Bret Sellman
- Microbiome Discovery, Microbial Sciences, BioPharmaceuticals R & D, AstraZeneca, Gaithersburg, MD 20878, USA;
| | - Willem J. B. van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center Rotterdam, 3015 Rotterdam, The Netherlands;
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Diseases Institute, University of Antwerp, 2610 Antwerp, Belgium; (J.V.); (L.T.)
- Correspondence: ; Tel.: +32-3-265-27-52
| |
Collapse
|
36
|
Tran VG, Venkatasubramaniam A, Adhikari RP, Krishnan S, Wang X, Le VTM, Le HN, Vu TTT, Schneider-Smith E, Aman MJ, Diep BA. Efficacy of Active Immunization With Attenuated α-Hemolysin and Panton-Valentine Leukocidin in a Rabbit Model of Staphylococcus aureus Necrotizing Pneumonia. J Infect Dis 2020; 221:267-275. [PMID: 31504652 DOI: 10.1093/infdis/jiz437] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus is a common pathogen causing infections in humans with various degrees of severity, with pneumonia being one of the most severe infections. In as much as staphylococcal pneumonia is a disease driven in large part by α-hemolysin (Hla) and Panton-Valentine leukocidin (PVL), we evaluated whether active immunization with attenuated forms of Hla (HlaH35L/H48L) alone, PVL components (LukS-PVT28F/K97A/S209A and LukF-PVK102A) alone, or combination of all 3 toxoids could prevent lethal challenge in a rabbit model of necrotizing pneumonia caused by the USA300 community-associated methicillin-resistant S. aureus (MRSA). Rabbits vaccinated with Hla toxoid alone or PVL components alone were only partially protected against lethal pneumonia, whereas those vaccinated with all 3 toxoids had 100% protection against lethality. Vaccine-mediated protection correlated with induction of polyclonal antibody response that neutralized not only α-hemolysin and PVL, but also other related toxins, produced by USA300 and other epidemic MRSA clones.
Collapse
Affiliation(s)
- Vuvi G Tran
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| | | | | | | | - Xing Wang
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| | - Vien T M Le
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| | - Hoan N Le
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| | - Trang T T Vu
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| | - Erika Schneider-Smith
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| | - M Javad Aman
- Integrated Biotherapeutics, Inc, Rockville, Maryland
| | - Binh An Diep
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco
| |
Collapse
|
37
|
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
|
38
|
Liu J, Kozhaya L, Torres VJ, Unutmaz D, Lu M. Structure-based discovery of a small-molecule inhibitor of methicillin-resistant Staphylococcus aureus virulence. J Biol Chem 2020; 295:5944-5959. [PMID: 32179646 PMCID: PMC7196633 DOI: 10.1074/jbc.ra120.012697] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Indexed: 01/07/2023] Open
Abstract
The rapid emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) strains poses a major threat to public health. MRSA possesses an arsenal of secreted host-damaging virulence factors that mediate pathogenicity and blunt immune defenses. Panton-Valentine leukocidin (PVL) and α-toxin are exotoxins that create lytic pores in the host cell membrane. They are recognized as being important for the development of invasive MRSA infections and are thus potential targets for antivirulence therapies. Here, we report the high-resolution X-ray crystal structures of both PVL and α-toxin in their soluble, monomeric, and oligomeric membrane-inserted pore states in complex with n-tetradecylphosphocholine (C14PC). The structures revealed two evolutionarily conserved phosphatidylcholine-binding mechanisms and their roles in modulating host cell attachment, oligomer assembly, and membrane perforation. Moreover, we demonstrate that the soluble C14PC compound protects primary human immune cells in vitro against cytolysis by PVL and α-toxin and hence may serve as the basis for the development of an antivirulence agent for managing MRSA infections.
Collapse
Affiliation(s)
- Jie Liu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Lina Kozhaya
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Min Lu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, To whom correspondence should be addressed:
Public Health Research Institute, Dept. of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Newark, NJ 07103. Tel.:
973-854-3260; E-mail:
| |
Collapse
|
39
|
Suppression of Staphylococcus aureus Superantigen-Independent Interferon Gamma Response by a Probiotic Polysaccharide. Infect Immun 2020; 88:IAI.00661-19. [PMID: 31932326 DOI: 10.1128/iai.00661-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive opportunistic pathogen that causes a variety of diseases. Bloodstream infection is the most severe, with mortality rates reaching 20 to 50%. Exopolysaccharide (EPS) from the probiotic Bacillus subtilis reduces bacterial burden and inflammation during S. aureus bloodstream infection in mice. Protection is due, in part, to hybrid macrophages that restrict S. aureus growth through reactive oxygen species and to limiting superantigen-induced T cell activation and interferon gamma (IFN-γ) production during infection. A decrease in IFN-γ production was observed within 24 h after infection, and here, we investigated how EPS abrogates its production. We discovered that S. aureus uses a rapid, superantigen-independent mechanism to induce host IFN-γ and that this is mediated by interleukin-12 (IL-12) activation of NK cells. Furthermore, we found that EPS limits IFN-γ production by modulating host immunity in a Toll-like receptor 4 (TLR4)-dependent manner, a signaling pathway that is required for EPS-mediated protection from S. aureus infection in vivo We conclude that EPS protects hosts from acute bloodstream S. aureus infection not only by inducing macrophages that restrict S. aureus growth and inhibit superantigen-activated T cells but also by limiting NK cell production of IFN-γ after S. aureus infection in a TLR4-dependent manner.
Collapse
|
40
|
Marchetti M, De Bei O, Bettati S, Campanini B, Kovachka S, Gianquinto E, Spyrakis F, Ronda L. Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development. Int J Mol Sci 2020; 21:E2145. [PMID: 32245010 PMCID: PMC7139808 DOI: 10.3390/ijms21062145] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Nutritional immunity is a form of innate immunity widespread in both vertebrates and invertebrates. The term refers to a rich repertoire of mechanisms set up by the host to inhibit bacterial proliferation by sequestering trace minerals (mainly iron, but also zinc and manganese). This strategy, selected by evolution, represents an effective front-line defense against pathogens and has thus inspired the exploitation of iron restriction in the development of innovative antimicrobials or enhancers of antimicrobial therapy. This review focuses on the mechanisms of nutritional immunity, the strategies adopted by opportunistic human pathogen Staphylococcus aureus to circumvent it, and the impact of deletion mutants on the fitness, infectivity, and persistence inside the host. This information finally converges in an overview of the current development of inhibitors targeting the different stages of iron uptake, an as-yet unexploited target in the field of antistaphylococcal drug discovery.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
| | - Omar De Bei
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Stefano Bettati
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Sandra Kovachka
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Luca Ronda
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
41
|
Yang H, Xu S, Huang K, Xu X, Hu F, He C, Shu W, Wang Z, Gong F, Zhang C, Liu Q. Anti-staphylococcus Antibiotics Interfere With the Transcription of Leucocidin ED Gene in Staphylococcus aureus Strain Newman. Front Microbiol 2020; 11:265. [PMID: 32194524 PMCID: PMC7066085 DOI: 10.3389/fmicb.2020.00265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 02/05/2020] [Indexed: 11/13/2022] Open
Abstract
Antibiotics have been described to modulate bacterial virulence gene expression. This study aimed to assess the changes caused by anti-Staphylococcus agents in the transcription of leucocidin ED (lukED) gene of Staphylococcus aureus strain Newman in vitro and in vivo and to determine whether the altered expression is agr dependent. The bacteria were exposed to subinhibitory concentrations [1/2, 1/4, or 1/8 minimal inhibitory concentration (MIC)] of 11 antibiotics, and the expression of lukE and agr-effector RNAIII was determined using qRT-PCR. In vivo experiments were performed to evaluate the impact exerted by six representative antibiotics on the transcription of both genes. Molecular analysis showed that in vitro lukE transcription was dramatically promoted in the Newman strain exposed to sub-MICs of vancomycin, trimethoprim-sulfamethoxazole, clindamycin, gentamicin, daptomycin, and ciprofloxacin and considerably reduced when stimulated by cefazolin, erythromycin, rifampicin, tigecycline, and linezolid. In the murine abscess model, tigecycline significantly decreased the transcription of lukE and the bacterial numbers, whereas vancomycin increased them; although cefazolin increased the lukE expression (contrary to the in vitro effect), it had a remarkable role in reducing bacterial load. The correspondence analysis shows that RNAIII expression varied under seven of 11 antibiotics in vitro, and six drugs in vivo were consistent with lukE transcripts. In conclusion, our data show that anti-Staphylococcus antibiotics exert modulatory effects on lukE expression in vitro and/or in vivo, and the changed expression caused by some drugs may be involved with agr activity, thus providing a guide to choose appropriate agents to avoid promoting bacterial virulence in lukED-positive S. aureus infections.
Collapse
Affiliation(s)
- Han Yang
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Su Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Kaifeng Huang
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaogang Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Fupin Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Chunyan He
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Shu
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyan Wang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Gong
- Department of Clinical Laboratory, The Third Hospital Affiliated to Nantong University, Wuxi, China
| | - Chuanling Zhang
- Department of Clinical Laboratory, Xiaoshan Hospital, Hangzhou, China
| | - Qingzhong Liu
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Chan R, Buckley PT, O'Malley A, Sause WE, Alonzo F, Lubkin A, Boguslawski KM, Payne A, Fernandez J, Strohl WR, Whitaker B, Lynch AS, Torres VJ. Identification of biologic agents to neutralize the bicomponent leukocidins of Staphylococcus aureus. Sci Transl Med 2020; 11:11/475/eaat0882. [PMID: 30651319 DOI: 10.1126/scitranslmed.aat0882] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 01/22/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022]
Abstract
A key aspect underlying the severity of infections caused by Staphylococcus aureus is the abundance of virulence factors that the pathogen uses to thwart critical components of the human immune response. One such mechanism involves the destruction of host immune cells by cytolytic toxins secreted by S. aureus, including five bicomponent leukocidins: PVL, HlgAB, HlgCB, LukED, and LukAB. Purified leukocidins can lyse immune cells ex vivo, and systemic injections of purified LukED or HlgAB can acutely kill mice. Here, we describe the generation and characterization of centyrins that bind S. aureus leukocidins with high affinity and protect primary human immune cells from toxin-mediated cytolysis. Centyrins are small protein scaffolds derived from the fibronectin type III-binding domain of the human protein tenascin-C. Although centyrins are potent in tissue culture assays, their short serum half-lives limit their efficacies in vivo. By extending the serum half-lives of centyrins through their fusion to an albumin-binding consensus domain, we demonstrate the in vivo efficacy of these biologics in a murine intoxication model and in models of both prophylactic and therapeutic treatment of live S. aureus systemic infections. These biologics that target S. aureus virulence factors have potential for treating and preventing serious staphylococcal infections.
Collapse
Affiliation(s)
- Rita Chan
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Peter T Buckley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - William E Sause
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Angela Payne
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Jeffrey Fernandez
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - William R Strohl
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Brian Whitaker
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Anthony Simon Lynch
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA.
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
43
|
Kläui AJ, Boss R, Graber HU. Characterization and Comparative Analysis of the Staphylococcus aureus Genomic Island vSaβ: an In Silico Approach. J Bacteriol 2019; 201:e00777-18. [PMID: 31451542 PMCID: PMC6805111 DOI: 10.1128/jb.00777-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/25/2019] [Indexed: 01/10/2023] Open
Abstract
Staphylococcus aureus is a widespread opportunistic pathogen to humans and animals. Of its genome, 20 to 25% varies between strains and consists of phages, pathogenicity islands, transposons, and genomic islands. S. aureus harbors up to three genomic islands, vSaα, vSaβ, and vSaγ. The vSaβ region of S. aureus can encode a number of virulence-associated factors, such as serine proteases, leukocidins, enterotoxins, bacteriocins, or a hyaluronate lyase. In this study, the vSaβ regions of 103 clinically relevant S. aureus strains were characterized in silico and compared to the three predefined vSaβ types. We here suggest a superordinate system of 15 different vSaβ types, of which 12 were newly defined. Each vSaβ type has a distinct structure with a distinct set of genes, which are both highly conserved. Between the different types, gene content and composition vary substantially. Based on our data, a strain's vSaβ type is strongly coupled with its clonal complex, suggesting that vSaβ was acquired in an ancestral S. aureus strain, arguably by phage mediation, before differentiation into clonal complexes. In addition, we addressed the issue of ambiguous nomenclature in the serine protease gene cluster and propose a novel, phylogeny-based nomenclature of the cluster contained in the vSaβ region.IMPORTANCE With the rapid increase of available sequencing data on clinically relevant bacterial species such as S. aureus, the genomic basis of clinical phenotypes can be investigated in much more detail, allowing a much deeper understanding of the mechanisms involved in disease. We characterized in detail the S. aureus genomic island vSaβ and defined a superordinate system to categorize S. aureus strains based on their vSaβ type, providing information about the strains' virulence-associated genes and clinical potential.
Collapse
Affiliation(s)
- Anita J Kläui
- Food Microbial Systems, Agroscope, Bern, Switzerland
| | - Renate Boss
- Risk Assessment Division, Federal Food Safety and Veterinary Office, Bern, Switzerland
| | - Hans U Graber
- Food Microbial Systems, Agroscope, Bern, Switzerland
| |
Collapse
|
44
|
Talley S, Kalinina O, Winek M, Paik W, Cannon AR, Alonzo F, Choudhry MA, Knight KL, Campbell EM. A Caspase-1 Biosensor to Monitor the Progression of Inflammation In Vivo. THE JOURNAL OF IMMUNOLOGY 2019; 203:2497-2507. [DOI: 10.4049/jimmunol.1900619] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/21/2019] [Indexed: 12/22/2022]
|
45
|
Keogh RA, Zapf RL, Trzeciak E, Null GG, Wiemels RE, Carroll RK. Novel Regulation of Alpha-Toxin and the Phenol-Soluble Modulins by Peptidyl-Prolyl cis/trans Isomerase Enzymes in Staphylococcus aureus. Toxins (Basel) 2019; 11:toxins11060343. [PMID: 31208155 PMCID: PMC6628628 DOI: 10.3390/toxins11060343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases) are enzymes that catalyze the cis-to-trans isomerization around proline bonds, allowing proteins to fold into their correct confirmation. Previously, we identified two PPIase enzymes in Staphylococcus aureus (PpiB and PrsA) that are involved in the regulation of virulence determinants and have shown that PpiB contributes to S. aureus virulence in a murine abscess model of infection. Here, we further examine the role of these PPIases in S. aureus virulence and, in particular, their regulation of hemolytic toxins. Using murine abscess and systemic models of infection, we show that a ppiB mutant in a USA300 background is attenuated for virulence but that a prsA mutant is not. Deletion of the ppiB gene leads to decreased bacterial survival in macrophages and nasal epithelial cells, while there is no significant difference when prsA is deleted. Analysis of culture supernatants reveals that a ppiB mutant strain has reduced levels of the phenol-soluble modulins and that both ppiB and prsA mutants have reduced alpha-toxin activity. Finally, we perform immunoprecipitation to identify cellular targets of PpiB and PrsA. Results suggest a novel role for PpiB in S. aureus protein secretion. Collectively, our results demonstrate that PpiB and PrsA influence S. aureus toxins via distinct mechanisms, and that PpiB but not PrsA contributes to disease.
Collapse
Affiliation(s)
- Rebecca A Keogh
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Rachel L Zapf
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Emily Trzeciak
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Gillian G Null
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Richard E Wiemels
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Ronan K Carroll
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
- The Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
46
|
Abstract
There are a plethora of probiotic formulae that supposedly benefit human health on the market. However, the scientific underpinnings of the claimed benefits have remained poorly established. Scientific evidence is now increasingly being provided that explains those benefits, for example, by immune-stimulatory effects or inter-bacterial competition between beneficial and pathogenic bacteria. In our recent study (Piewngam et al. Nature 2018), we show that Bacillus colonization of the human intestine is negatively correlated with that of the human pathogen, Staphylococcus aureus. This type of colonization resistance is achieved by secretion of a class of lipopeptides by Bacillus species that inhibits S. aureus quorum-sensing signaling, which we found is crucial for S. aureus intestinal colonization. Here, we discuss what these findings imply for the general role of S. aureus intestinal colonization, the role of quorum-sensing in that process, and potential alternative ways to control S. aureus infection.
Collapse
Affiliation(s)
- Pipat Piewngam
- Pathogen Molecular Genetics Section, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, MD, USA,CONTACT Michael Otto Pathogen Molecular Genetics Section, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, 50 South Drive, Bethesda, MD 20814
| |
Collapse
|
47
|
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
|
48
|
Cosgriff CJ, White CR, Teoh WP, Grayczyk JP, Alonzo F. Control of Staphylococcus aureus Quorum Sensing by a Membrane-Embedded Peptidase. Infect Immun 2019; 87:e00019-19. [PMID: 30833334 PMCID: PMC6479040 DOI: 10.1128/iai.00019-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/23/2019] [Indexed: 02/08/2023] Open
Abstract
Gram-positive bacteria process and release small peptides, or pheromones, that act as signals for the induction of adaptive traits, including those involved in pathogenesis. One class of small signaling pheromones is the cyclic autoinducing peptides (AIPs), which regulate expression of genes that orchestrate virulence and persistence in a range of microbes, including staphylococci, listeriae, clostridia, and enterococci. In a genetic screen for Staphylococcus aureus secreted virulence factors, we identified an S. aureus mutant containing an insertion in the gene SAUSA300_1984 (mroQ), which encodes a putative membrane-embedded metalloprotease. A ΔmroQ mutant exhibited impaired induction of Toll-like receptor 2-dependent inflammatory responses from macrophages but elicited greater production of the inflammatory cytokine interleukin-1β and was attenuated in a murine skin and soft tissue infection model. The ΔmroQ mutant phenocopies an S. aureus mutant containing a deletion of the accessory gene regulatory system (Agr), wherein both strains have significantly reduced production of secreted toxins and virulence factors but increased surface protein A abundance. The Agr system controls virulence factor gene expression in S. aureus by sensing the accumulation of AIP via the histidine kinase AgrC and the response regulator AgrA. We provide evidence to suggest that MroQ acts within the Agr pathway to facilitate the optimal processing or export of AIP for signal amplification through AgrC/A and induction of virulence factor gene expression. Mutation of MroQ active-site residues significantly reduces AIP signaling and attenuates virulence. Altogether, this work identifies a new component of the Agr quorum-sensing circuit that is critical for the production of S. aureus virulence factors.
Collapse
Affiliation(s)
- Chance J Cosgriff
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Chelsea R White
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Wei Ping Teoh
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - James P Grayczyk
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Francis Alonzo
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| |
Collapse
|
49
|
Lubkin A, Lee WL, Alonzo F, Wang C, Aligo J, Keller M, Girgis NM, Reyes-Robles T, Chan R, O'Malley A, Buckley P, Vozhilla N, Vasquez MT, Su J, Sugiyama M, Yeung ST, Coffre M, Bajwa S, Chen E, Martin P, Kim SY, Loomis C, Worthen GS, Shopsin B, Khanna KM, Weinstock D, Lynch AS, Koralov SB, Loke P, Cadwell K, Torres VJ. Staphylococcus aureus Leukocidins Target Endothelial DARC to Cause Lethality in Mice. Cell Host Microbe 2019; 25:463-470.e9. [PMID: 30799265 DOI: 10.1016/j.chom.2019.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/24/2018] [Accepted: 01/23/2019] [Indexed: 01/16/2023]
Abstract
The pathogenesis of Staphylococcus aureus is thought to depend on the production of pore-forming leukocidins that kill leukocytes and lyse erythrocytes. Two leukocidins, Leukocidin ED (LukED) and γ-Hemolysin AB (HlgAB), are necessary and sufficient to kill mice upon infection and toxin challenge. We demonstrate that LukED and HlgAB cause vascular congestion and derangements in vascular fluid distribution that rapidly cause death in mice. The Duffy antigen receptor for chemokines (DARC) on endothelial cells, rather than leukocytes or erythrocytes, is the critical target for lethality. Consistent with this, LukED and HlgAB injure primary human endothelial cells in a DARC-dependent manner, and mice with DARC-deficient endothelial cells are resistant to toxin-mediated lethality. During bloodstream infection in mice, DARC targeting by S. aureus causes increased tissue damage, organ dysfunction, and host death. The potential for S. aureus leukocidins to manipulate vascular integrity highlights the importance of these virulence factors.
Collapse
Affiliation(s)
- Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Warren L Lee
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Changsen Wang
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Jason Aligo
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Matthew Keller
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Natasha M Girgis
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Tamara Reyes-Robles
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Rita Chan
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Peter Buckley
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Nikollaq Vozhilla
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Marilyn T Vasquez
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Johnny Su
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Michael Sugiyama
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Stephen T Yeung
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Maryaline Coffre
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Sofia Bajwa
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Eric Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Patricia Martin
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Sang Y Kim
- Department of Pathology, New York University School of Medicine, New York, NY, USA; Office of Collaborative Sciences, NYU School of Medicine, New York, NY, USA; Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Cynthia Loomis
- Department of Pathology, New York University School of Medicine, New York, NY, USA; Office of Collaborative Sciences, NYU School of Medicine, New York, NY, USA; Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - G Scott Worthen
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bo Shopsin
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; Division of Infectious Diseases, Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Daniel Weinstock
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Anthony Simon Lynch
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - P'ng Loke
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Ken Cadwell
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
50
|
Adaptation of the Staphylococcus aureus leukocidin LukGH for the rabbit host by protein engineering. Biochem J 2019; 476:275-292. [PMID: 30559327 DOI: 10.1042/bcj20180691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023]
Abstract
Host defense against Staphylococcus aureus greatly depends on bacterial clearance by phagocytic cells. LukGH (or LukAB) is the most potent staphylococcal leukocidin towards human phagocytes in vitro, but its role in pathogenesis is obscured by the lack of suitable small animal models because LukGH has limited or no cytotoxicity towards rodent and rabbit compared with human polymorphonuclear cells (PMNs) likely due to an impaired interaction with its cellular receptor, CD11b. We aimed at adapting LukGH for the rabbit host by improving binding to the rabbit homolog of CD11b, specifically its I-domain (CD11b-I). Targeted amino acid substitutions were introduced into the LukH polypeptide to map its receptor interaction site(s). We found that the binding affinity of LukGH variants to the human and rabbit CD11b-I correlated well with their PMN cytotoxicity. Importantly, we identified LukGH variants with significantly improved cytotoxicity towards rabbit PMNs, when expressed recombinantly (10-15-fold) or by engineered S. aureus strains. These findings support the development of small animal models of S. aureus infection with the potential for demonstrating the importance of LukGH in pathogenesis.
Collapse
|