1
|
Dufresne K, Tuffs SW, Walton NR, Kasper KJ, Mohorovic I, Hasan F, Bentall T, Heinrichs DE, Delport J, Mele TS, McCormick JK. Evidence that staphylococcal superantigens promote within-patient bacterial persistence following post-operative surgical site infection. Infect Immun 2025; 93:e0040724. [PMID: 39878522 PMCID: PMC11895439 DOI: 10.1128/iai.00407-24] [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: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Staphylococcus aureus is a predominant cause of post-operative surgical site infections and persistent bacteremia. Here, we describe a patient who experienced three episodes of S. aureus infection over a period of 4 months following a total knee arthroplasty. The initial bloodstream isolate (SAB-0429) was a clonal complex 5 (CC5) and methicillin-resistant S. aureus (MRSA), whereas two subsequent isolates (SAB-0485 and SAB-0495) were CC5 isolates but methicillin-sensitive S. aureus. The two latter isolates harbored a plasmid encoding three superantigen genes that were not present in the primary MRSA isolate. SAB-0485 and SAB-0495 both expressed the plasmid-encoded staphylococcal enterotoxin R exotoxin and demonstrated increased superantigen activity compared with SAB-0429. Compared to SAB-0429, the latter isolates also demonstrated an increased bacterial burden in a mouse bacteremia model that was dependent on increased interferon-γ production. Curing of the plasmid from SAB-0485 reduced this virulence phenotype. These findings suggest that the superantigen exotoxins may provide a selective advantage in chronic post-surgical infections.IMPORTANCEIn this study, we investigated bacterial isolates from a patient who experienced three recurrent S. aureus infections over a 4 month period following total knee arthroplasty. Genomic and phenotypic characterization of these isolates revealed that they all belonged to clonal complex 5, yet the latter two strains contained an additional plasmid encoding superantigen exotoxins. Subsequent experimental infection experiments in mice demonstrated that the plasmid-encoded superantigens exacerbated bacteremia by promoting liver abscess formation. These experiments suggest that despite appropriate antibiotic therapy, bacterial superantigens may be able to promote persistent infection following post-surgery.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Stephen W. Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Nicholas R. Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Katherine J. Kasper
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Ivor Mohorovic
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Farah Hasan
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tracey Bentall
- Division of Critical Care Medicine, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - David E. Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Johan Delport
- Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | - Tina S. Mele
- Division of Critical Care Medicine, Department of Medicine, University of Western Ontario, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, London Health Sciences Centre, University Hospital, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
2
|
Zhu Y, Luo J, Xia X, Feng H, Zhao P. Differential neutrophil responses in murine following intraperitoneal injections of Escherichia coli and Staphylococcus aureus. Heliyon 2024; 10:e40281. [PMID: 39641065 PMCID: PMC11617748 DOI: 10.1016/j.heliyon.2024.e40281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/13/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Objective This study aimed to investigate the proportion of neutrophils among leukocytes, in various tissues following intraperitoneal injection of Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) in mice. Methods Twelve specific-pathogen free (SPF) male mice, aged eight weeks, were segregated into three groups, each containing four mice. Two of these groups were subjected to intraperitoneal injections of E. coli and S. aureus, both in high concentrations, to establish mouse models of inflammation. The remaining group, which received an intraperitoneal injection of phosphate buffered saline (PBS), served as the control group. Observe the mice every half hour. Then mice were anesthetized, and samples from peripheral blood, liver, and brain tissues were carefully collected nearing death. These samples underwent a digestion process to produce single-cell suspensions. Subsequently, these suspensions were stained with fluorescent antibodies targeting CD45, Ly6G, and CD11b. A flow cytometric analyzer was then employed to enumerate and compare the neutrophil alterations across each group (Fig. 1). Results The results indicated a significant variation in the ratio of CD11b+ Ly6G+ neutrophils to CD45+ leukocytes among the groups. In peripheral blood, the control group showed a neutrophil proportion of approximately 1.44 %, while the E. coli and S. aureus groups exhibited increased proportions of 6.53 % and 3.82 %, respectively. In liver tissue, a marked elevation was observed in the experimental groups, with ratios of 19.20 % and 20.40 % for E. coli and S. aureus, respectively, compared to 1.64 % in the control. In brain tissue, the increments were more modest but noticeable, with the experimental groups showing 2.40 % and 1.11 % in contrast to 0.13 % in the control group. Conclusions These findings suggest neutrophils are involved in the response after intraperitoneal injection of E. coli and S. aureus, with marked differences in neutrophil responses in different tissues. This study enhances our understanding of the acute inflammatory response to bacterial infection.
Collapse
Affiliation(s)
- Yanyan Zhu
- Department of Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-Quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, China
| | - Jingya Luo
- Department of Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-Quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xianzhu Xia
- Department of Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-Quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Hao Feng
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, China
| | - Pingsen Zhao
- Department of Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-Quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| |
Collapse
|
3
|
Zhang H, Monk IR, Braverman J, Jones CM, Brooks AG, Stinear TP, Wakim LM. Staphylococcal superantigens evoke temporary and reversible T cell anergy, but fail to block the development of a bacterium specific cellular immune response. Nat Commun 2024; 15:9872. [PMID: 39543088 PMCID: PMC11564628 DOI: 10.1038/s41467-024-54074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Superantigens (sAgs) are bacterial virulence factors that induce a state of immune hyperactivation by forming a bridge between certain subsets of T cell receptor (TCR) β chains on T lymphocytes, and class II major histocompatibility complex (MHC-II) molecules; this cross-linking leads to indiscriminate T cell activation, cytokine storm and toxic shock. Here we show that sAg exposure drives the preferential expansion of naive and central memory T cell subsets, but not effector or resident memory T cells, which instead, hyper release pro-inflammatory cytokines. A targeted therapeutic approach to minimise cytokine release by effector memory T cells attenuated sAg-induced cytokine release. Irrespective of antigen experience, sAg activation does not render mature T cells permanently dysfunctional, and full restoration of effector function is observed following a transient and reversible anergy. Moreover, we show that in the face of sAg induced immune hyperactivation, an intact bacterium-specific CD4+ T cell response can be mounted.
Collapse
Affiliation(s)
- Heran Zhang
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Ian R Monk
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Jessica Braverman
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Claerwen M Jones
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
4
|
Tuffs SW, Dufresne K, Rishi A, Walton NR, McCormick JK. Novel insights into the immune response to bacterial T cell superantigens. Nat Rev Immunol 2024; 24:417-434. [PMID: 38225276 DOI: 10.1038/s41577-023-00979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Bacterial T cell superantigens (SAgs) are a family of microbial exotoxins that function to activate large numbers of T cells simultaneously. SAgs activate T cells by direct binding and crosslinking of the lateral regions of MHC class II molecules on antigen-presenting cells with T cell receptors (TCRs) on T cells; these interactions alter the normal TCR-peptide-MHC class II architecture to activate T cells in a manner that is independent of the antigen specificity of the TCR. SAgs have well-recognized, central roles in human diseases such as toxic shock syndrome and scarlet fever through their quantitative effects on the T cell response; in addition, numerous other consequences of SAg-driven T cell activation are now being recognized, including direct roles in the pathogenesis of endocarditis, bloodstream infections, skin disease and pharyngitis. In this Review, we summarize the expanding family of bacterial SAgs and how these toxins can engage highly diverse adaptive immune receptors. We highlight recent findings regarding how SAg-driven manipulation of the adaptive immune response may operate in multiple human diseases, as well as contributing to the biology and life cycle of SAg-producing bacterial pathogens.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Aanchal Rishi
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Nicholas R Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - John K McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
5
|
Santacroce L, Topi S, Charitos IA, Lovero R, Luperto P, Palmirotta R, Jirillo E. Current Views about the Inflammatory Damage Triggered by Bacterial Superantigens and Experimental Attempts to Neutralize Superantigen-Mediated Toxic Effects with Natural and Biological Products. PATHOPHYSIOLOGY 2024; 31:18-31. [PMID: 38251046 PMCID: PMC10801599 DOI: 10.3390/pathophysiology31010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/23/2024] Open
Abstract
Superantigens, i.e., staphylococcal enterotoxins and toxic shock syndrome toxin-1, interact with T cells in a different manner in comparison to conventional antigens. In fact, they activate a larger contingent of T lymphocytes, binding outside the peptide-binding groove of the major histocompatibility complex class II. Involvement of many T cells by superantigens leads to a massive release of pro-inflammatory cytokines, such as interleukin (IL)-1, IL-2, IL-6, tumor necrosis factor-alpha and interferon-gamma. Such a storm of mediators has been shown to account for tissue damage, multiorgan failure and shock. Besides conventional drugs and biotherapeutics, experiments with natural and biological products have been undertaken to attenuate the toxic effects exerted by superantigens. In this review, emphasis will be placed on polyphenols, probiotics, beta-glucans and antimicrobial peptides. In fact, these substances share a common functional denominator, since they skew the immune response toward an anti-inflammatory profile, thus mitigating the cytokine wave evoked by superantigens. However, clinical applications of these products are still scarce, and more trials are needed to validate their usefulness in humans.
Collapse
Affiliation(s)
- Luigi Santacroce
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Skender Topi
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania
| | - Ioannis Alexandros Charitos
- Division of Pneumology and Respiratory Rehabilitation, Maugeri Clinical Scientific Research Institutes (IRCCS) of Pavia—Scientific Institute of Bari, 70124 Bari, Italy
| | - Roberto Lovero
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | | | - Raffaele Palmirotta
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Emilio Jirillo
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| |
Collapse
|
6
|
Hullahalli K, Dailey KG, Hasegawa Y, Torres E, Suzuki M, Zhang H, Threadgill DW, Navarro VM, Waldor MK. Genetic and immune determinants of E. coli liver abscess formation. Proc Natl Acad Sci U S A 2023; 120:e2310053120. [PMID: 38096412 PMCID: PMC10743367 DOI: 10.1073/pnas.2310053120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Systemic infections can yield distinct outcomes in different tissues. In mice, intravenous inoculation of Escherichia coli leads to bacterial replication within liver abscesses, while other organs such as the spleen clear the pathogen. Abscesses are macroscopic necrotic regions that comprise the vast majority of the bacterial burden in the animal, yet little is known about the processes underlying their formation. Here, we characterize E. coli liver abscesses and identify host determinants of abscess susceptibility. Spatial transcriptomics revealed that liver abscesses are associated with heterogenous immune cell clusters comprised of macrophages, neutrophils, dendritic cells, innate lymphoid cells, and T-cells that surround necrotic regions of the liver. Abscess susceptibility is heightened in the C57BL lineage, particularly in C57BL/6N females. Backcross analyses demonstrated that abscess susceptibility is a polygenic trait inherited in a sex-dependent manner without direct linkage to sex chromosomes. As early as 1 d post infection, the magnitude of E. coli replication in the liver distinguishes abscess-susceptible and abscess-resistant strains of mice, suggesting that the immune pathways that regulate abscess formation are induced within hours. We characterized the early hepatic response with single-cell RNA sequencing and found that mice with reduced activation of early inflammatory responses, such as those lacking the LPS receptor TLR4 (Toll-like receptor 4), are resistant to abscess formation. Experiments with barcoded E. coli revealed that TLR4 mediates a tradeoff between abscess formation and bacterial clearance. Together, our findings define hallmarks of E. coli liver abscess formation and suggest that hyperactivation of the hepatic innate immune response drives liver abscess susceptibility.
Collapse
Affiliation(s)
- Karthik Hullahalli
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA02115
| | - Katherine G. Dailey
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA02115
| | - Yuko Hasegawa
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA02115
| | - Encarnacion Torres
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Masataka Suzuki
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA02115
| | - Hailong Zhang
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA02115
| | - David W. Threadgill
- Department of Cell Biology and Genetics, Texas A&M University, College Station, TX76549
- Department of Nutrition, Texas A&M University, College Station, TX76549
| | - Victor M. Navarro
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Matthew K. Waldor
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA02115
| |
Collapse
|
7
|
Li F, Niu B, Liu L, Zhu M, Yang H, Qin B, Peng X, Chen L, Xu C, Zhou X. Characterization of genetic humanized mice with transgenic HLA DP401 or DRA but deficient in endogenous murine MHC class II genes upon Staphylococcus aureus pneumonia. Animal Model Exp Med 2023; 6:585-597. [PMID: 37246733 PMCID: PMC10757210 DOI: 10.1002/ame2.12331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 05/09/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Staphylococcus aureus can cause serious infections by secreting many superantigen exotoxins in "carrier" or "pathogenic" states. HLA DQ and HLA DR humanized mice have been used as a small animal model to study the role of two molecules during S. aureus infection. However, the contribution of HLA DP to S. aureus infection is unknown yet. METHODS In this study, we have produced HLA DP401 and HLA DRA0101 humanized mice by microinjection of C57BL/6J zygotes. Neo-floxed IAβ+/- mice were crossbred with Ella-Cre and further crossbred with HLA DP401 or HLA-DRA0101 humanized mice. After several rounds of traditional crossbreeding, we finally obtained HLA DP401-IAβ-/- and HLA DRA-IAβ-/- humanized mice, in which human DP401 or DRA0101 molecule was introduced into IAβ-/- mice deficient in endogenous murine MHC class II molecules. A transnasal infection murine model of S. aureus pneumonia was induced in the humanized mice by administering 2 × 108 CFU of S. aureus Newman dropwise into the nasal cavity. The immune responses and histopathology changes were further assessed in lungs in these infected mice. RESULTS We evaluated the local and systemic effects of S. aureus delivered intranasally in HLA DP401-IAβ-/- and HLA DRA-IAβ-/- transgenic mice. S. aureus Newman infection significantly increased the mRNA level of IL 12p40 in lungs in humanized mice. An increase in IFN-γ and IL-6 protein was observed in HLA DRA-IAβ-/- mice. We observed a declining trend in the percentage of F4/80+ macrophages in lungs in HLA DP401-IAβ-/- mice and a decreasing ratio of CD4+ to CD8+ T cells in lungs in IAβ-/- mice and HLA DP401-IAβ-/- mice. A decreasing ratio of Vβ3+ to Vβ8+ T cells was also found in the lymph node of IAβ-/- mice and HLA DP401-IAβ-/- mice. S. aureus Newman infection resulted in a weaker pathological injury in lungs in IAβ-/- genetic background mice. CONCLUSION These humanized mice will be an invaluable mouse model to resolve the pathological mechanism of S. aureus pneumonia and study what role DP molecule plays in S. aureus infection.
Collapse
Affiliation(s)
- Feng Li
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Bowen Niu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Lingling Liu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Mengmin Zhu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Hua Yang
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Boyin Qin
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Xiuhua Peng
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Lixiang Chen
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Chunhua Xu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| | - Xiaohui Zhou
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterShanghaiChina
| |
Collapse
|
8
|
Hullahalli K, Dailey KG, Hasegawa Y, Suzuki M, Zhang H, Threadgill DW, Waldor MK. Genetic and immune determinants of E. coli liver abscess formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.11.543319. [PMID: 37398354 PMCID: PMC10312621 DOI: 10.1101/2023.06.11.543319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Systemic infections can yield distinct outcomes in different tissues. In mice, intravenous inoculation of E. coli leads to bacterial replication within liver abscesses while other organs such as the spleen largely clear the pathogen. Abscesses are macroscopic necrotic regions that comprise the vast majority of the bacterial burden in the animal, yet little is known about the processes underlying their formation. Here, we characterize E. coli liver abscesses and identify host determinants of abscess susceptibility. Spatial transcriptomics revealed that liver abscesses are associated with heterogenous immune cell clusters comprised of macrophages, neutrophils, dendritic cells, innate lymphoid cells, and T-cells that surround necrotic regions of the liver. Susceptibility to liver abscesses is heightened in the C57BL/6 lineage, particularly in C57BL/6N females. Backcross analyses demonstrated that abscess susceptibility is a polygenic trait inherited in a sex-dependent manner without direct linkage to sex chromosomes. As early as one day post infection, the magnitude of E. coli replication in the liver distinguishes abscess-susceptible and abscess-resistant strains of mice, suggesting that the immune pathways that regulate abscess formation are induced within hours. We characterized the early hepatic response with single-cell RNA sequencing and found that mice with reduced activation of early inflammatory responses, such as those lacking the LPS receptor TLR4, are resistant to abscess formation. Experiments with barcoded E. coli revealed that TLR4 mediates a tradeoff between abscess formation and bacterial clearance. Together, our findings define hallmarks of E. coli liver abscess formation and suggest that hyperactivation of the hepatic innate immune response drives liver abscess susceptibility.
Collapse
Affiliation(s)
- Karthik Hullahalli
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - Katherine G Dailey
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - Yuko Hasegawa
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - Masataka Suzuki
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - Hailong Zhang
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - David W Threadgill
- Department of Cell Biology and Genetics and Department of Nutrition, Texas A&M University, College Station, TX 76549, USA
| | - Matthew K Waldor
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| |
Collapse
|
9
|
Mikhaylova Y, Shelenkov A, Chernyshkov A, Tyumentseva M, Saenko S, Egorova A, Manzeniuk I, Akimkin V. Whole-Genome Analysis of Staphylococcus aureus Isolates from Ready-to-Eat Food in Russia. Foods 2022; 11:foods11172574. [PMID: 36076759 PMCID: PMC9455289 DOI: 10.3390/foods11172574] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
This study provides a thorough investigation of a diverse set of antimicrobial resistant (AMR) Staphylococcus aureus isolates collected from a broad range of ready-to-eat (RTE) food in various geographic regions of Russia ranging from Pskov to Kamchatka. Thirty-five isolates were characterized using the whole genome sequencing (WGS) analysis in terms of clonal structure, the presence of resistance and virulence determinants, as well as plasmid replicon sequences and CRISPR/Cas systems. To the best of our knowledge, this is the first WGS-based surveillance of Russian RTE food-associated S. aureus isolates. The isolates belonged to fifteen different multilocus sequence typing (MLST)-based types with a predominant being the ones of clonal complex (CC) 22. The isolates studied can pose a threat to public health since about 40% of the isolates carried at least one enterotoxin gene, and 70% of methicillin-resistant (MRSA) isolates carried a tsst1 gene encoding a toxin that may cause severe acute disease. In addition, plasmid analysis revealed some important characteristics, e.g., Rep5 and Rep20 plasmid replicons were a “signature” of MRSA CC22. By analyzing the isolates belonging to the same/single strain based on cgMLST analysis, we were able to identify the differences in their accessory genomes marking their dynamics and plasticity. This data is very important since S. aureus isolates studied and RTE food, in general, represent an important route of transmission and dissemination of multiple pathogenic determinants. We believe that the results obtained will facilitate performing epidemiological surveillance and developing protection measures against this important pathogen in community settings.
Collapse
|
10
|
Superantigens promote Staphylococcus aureus bloodstream infection by eliciting pathogenic interferon-gamma production. Proc Natl Acad Sci U S A 2022; 119:2115987119. [PMID: 35165181 PMCID: PMC8872782 DOI: 10.1073/pnas.2115987119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 12/29/2022] Open
Abstract
Since their discovery over 30 y ago, it has become clear that the superantigens (SAgs) are important virulence factors produced during severe Staphylococcus aureus–mediated disease including bacteremia. However, until the current study, it was unclear how these toxins manipulated the immune system to promote infection. Here, we have demonstrated that the SAgs can target a critical immune signaling molecule (interferon gamma), inducing overproduction that promotes bacterial survival by subverting the ability of macrophages to be able to kill the pathogen. This highlights SAg activity as a critical target for antistaphylococcal therapy to mitigate the impact of severe S. aureus disease. Staphylococcus aureus is a foremost bacterial pathogen responsible for a vast array of human diseases. Staphylococcal superantigens (SAgs) constitute a family of exotoxins from S. aureus that bind directly to major histocompatibility complex (MHC) class II and T cell receptors to drive extensive T cell activation and cytokine release. Although these toxins have been implicated in serious disease, including toxic shock syndrome, the specific pathological mechanisms remain unclear. Herein, we aimed to elucidate how SAgs contribute to pathogenesis during bloodstream infections and utilized transgenic mice encoding human MHC class II to render mice susceptible to SAg activity. We demonstrate that SAgs contribute to S. aureus bacteremia by massively increasing bacterial burden in the liver, and this was mediated by CD4+ T cells that produced interferon gamma (IFN-γ) to high levels in a SAg-dependent manner. Bacterial burdens were reduced by blocking IFN-γ, phenocopying SAg-deletion mutant strains, and inhibiting a proinflammatory response. Infection kinetics and flow cytometry analyses suggested that this was a macrophage-driven mechanism, which was confirmed through macrophage-depletion experiments. Experiments in human cells demonstrated that excessive IFN-γ allowed S. aureus to replicate efficiently within macrophages. This indicates that SAgs promote bacterial survival by manipulating the immune response to inhibit effective clearing of S. aureus. Altogether, this work implicates SAg toxins as critical therapeutic targets for preventing persistent or severe S. aureus disease.
Collapse
|
11
|
Braverman J, Monk IR, Ge C, Westall GP, Stinear TP, Wakim LM. Staphylococcus aureus specific lung resident memory CD4 + Th1 cells attenuate the severity of influenza virus induced secondary bacterial pneumonia. Mucosal Immunol 2022; 15:783-796. [PMID: 35637249 PMCID: PMC9148937 DOI: 10.1038/s41385-022-00529-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/25/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023]
Abstract
Staphylococcus aureus is a major cause of severe pulmonary infections. The evolution of multi-drug resistant strains limits antibiotic treatment options. To date, all candidate vaccines tested have failed, highlighting the need for an increased understanding of the immunological requirements for effective S. aureus immunity. Using an S. aureus strain engineered to express a trackable CD4+ T cell epitope and a murine model of S. aureus pneumonia, we show strategies that lodge Th1 polarised bacterium specific CD4+ tissue resident memory T cells (Trm) in the lung can significantly attenuate the severity of S. aureus pneumonia. This contrasts natural infection of mice that fails to lodge CD4+ Trm cells along the respiratory tract or provide protection against re-infection, despite initially generating Th17 bacterium specific CD4+ T cell responses. Interestingly, lack of CD4+ Trm formation after natural infection in mice appears to be reflected in humans, where the frequency of S. aureus reactive CD4+ Trm cells in lung tissue is also low. Our findings reveal the protective capacity of S. aureus specific respiratory tract CD4+ Th1 polarised Trm cells and highlight the potential for targeting these cells in vaccines that aim to prevent the development of S. aureus pneumonia.
Collapse
Affiliation(s)
- Jessica Braverman
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Ian R. Monk
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Chenghao Ge
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia ,grid.12527.330000 0001 0662 3178School of Medicine, Tsinghua University, Beijing, China
| | - Glen P. Westall
- grid.1002.30000 0004 1936 7857Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, VIC Australia
| | - Timothy P. Stinear
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
12
|
Côrtes MF, Botelho AMN, Bandeira PT, Mouton W, Badiou C, Bes M, Lima NCB, Soares AER, Souza RC, Almeida LGP, Martins-Simoes P, Vasconcelos ATR, Nicolás MF, Laurent F, Planet PJ, Figueiredo AMS. Reductive evolution of virulence repertoire to drive the divergence between community- and hospital-associated methicillin-resistant Staphylococcus aureus of the ST1 lineage. Virulence 2021; 12:951-967. [PMID: 33734031 PMCID: PMC7993186 DOI: 10.1080/21505594.2021.1899616] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 01/26/2021] [Accepted: 03/01/2021] [Indexed: 01/01/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) of the ST1-SCCmecIV lineage has been associated with community-acquired (CA) infections in North America and Australia. In Brazil, multi-drug resistant ST1-SCCmecIV MRSA has emerged in hospital-associated (HA) diseases in Rio de Janeiro. To understand these epidemiological differences, genomic and phylogenetic analyses were performed. In addition, virulence assays were done for representative CA - and HA-MRSA strains. Despite the conservation of the virulence repertoire, some genes were missing in Brazilian ST1-SCCmecIV including lukSF-PV, fnbB, and several superantigen-encoded genes. Additionally, CA-MRSA lost the splDE while HA-MRSA strains conserved the complete operon. Most of these variable genes were located in mobile genetic elements (MGE). However, conservation and maintenance of MGEs were often observed despite the absence of their associated virulence markers. A Bayesian phylogenetic tree revealed the occurrence of more than one entrance of ST1 strains in Rio de Janeiro. The tree shape and chronology allowed us to infer that the hospital-associated ST1-SCCmecIV from Brazil and the community-acquired USA400 from North America are not closely related and that they might have originated from different MSSA strains that independently acquired SCCmecIV cassettes. As expected, representatives of ST1 strains from Brazil showed lower cytotoxicity and a greater ability to survive inside human host cells. We suggest that Brazilian ST1-SCCmecIV strains have adapted to the hospital setting by reducing virulence and gaining the ability to persist and survive inside host cells. Possibly, these evolutionary strategies may balance the biologic cost of retaining multiple antibiotic resistance genes.
Collapse
Affiliation(s)
- Marina Farrel Côrtes
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Faculté de Médecine Lyon Est, Université de Lyon, Domaine de la Buire, Lyon, France
| | - Ana Maria N. Botelho
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Terra Bandeira
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - William Mouton
- Centre International de Recherche en Infectiologie (CIRI)―team Pathogénie des Staphylococques―Université Lyon 1, École Normale Supérieure de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils De Lyon, Lyon, France
| | - Cedric Badiou
- Centre International de Recherche en Infectiologie (CIRI)―team Pathogénie des Staphylococques―Université Lyon 1, École Normale Supérieure de Lyon, Lyon, France
| | - Michèle Bes
- Centre International de Recherche en Infectiologie (CIRI)―team Pathogénie des Staphylococques―Université Lyon 1, École Normale Supérieure de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils De Lyon, Lyon, France
- Laboratoire de Bactériologie, Centre de Biologie et de Pathologie Nord, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Nicholas C. B. Lima
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | | | - Rangel C. Souza
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Luiz G. P. Almeida
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Patricia Martins-Simoes
- Centre International de Recherche en Infectiologie (CIRI)―team Pathogénie des Staphylococques―Université Lyon 1, École Normale Supérieure de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils De Lyon, Lyon, France
- Laboratoire de Bactériologie, Centre de Biologie et de Pathologie Nord, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | | | - Marisa F. Nicolás
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Frédéric Laurent
- Faculté de Médecine Lyon Est, Université de Lyon, Domaine de la Buire, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI)―team Pathogénie des Staphylococques―Université Lyon 1, École Normale Supérieure de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils De Lyon, Lyon, France
- Laboratoire de Bactériologie, Centre de Biologie et de Pathologie Nord, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Paul J. Planet
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY, USA
| | - Agnes M. S. Figueiredo
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Hullahalli K, Waldor MK. Pathogen clonal expansion underlies multiorgan dissemination and organ-specific outcomes during murine systemic infection. eLife 2021; 10:e70910. [PMID: 34636322 PMCID: PMC8545400 DOI: 10.7554/elife.70910] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/07/2021] [Indexed: 12/31/2022] Open
Abstract
The dissemination of pathogens through blood and their establishment within organs lead to severe clinical outcomes. However, the within-host dynamics that underlie pathogen spread to and clearance from systemic organs remain largely uncharacterized. In animal models of infection, the observed pathogen population results from the combined contributions of bacterial replication, persistence, death, and dissemination, each of which can vary across organs. Quantifying the contribution of each these processes is required to interpret and understand experimental phenotypes. Here, we leveraged STAMPR, a new barcoding framework, to investigate the population dynamics of extraintestinal pathogenic Escherichia coli, a common cause of bacteremia, during murine systemic infection. We show that while bacteria are largely cleared by most organs, organ-specific clearance failures are pervasive and result from dramatic expansions of clones representing less than 0.0001% of the inoculum. Clonal expansion underlies the variability in bacterial burden between animals, and stochastic dissemination of clones profoundly alters the pathogen population structure within organs. Despite variable pathogen expansion events, host bottlenecks are consistent yet highly sensitive to infection variables, including inoculum size and macrophage depletion. We adapted our barcoding methodology to facilitate multiplexed validation of bacterial fitness determinants identified with transposon mutagenesis and confirmed the importance of bacterial hexose metabolism and cell envelope homeostasis pathways for organ-specific pathogen survival. Collectively, our findings provide a comprehensive map of the population biology that underlies bacterial systemic infection and a framework for barcode-based high-resolution mapping of infection dynamics.
Collapse
Affiliation(s)
- Karthik Hullahalli
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Division of Infectious Diseases, Brigham & Women’s HospitalBostonUnited States
| | - Matthew K Waldor
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Division of Infectious Diseases, Brigham & Women’s HospitalBostonUnited States
| |
Collapse
|
14
|
Deacy AM, Gan SKE, Derrick JP. Superantigen Recognition and Interactions: Functions, Mechanisms and Applications. Front Immunol 2021; 12:731845. [PMID: 34616400 PMCID: PMC8488440 DOI: 10.3389/fimmu.2021.731845] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Superantigens are unconventional antigens which recognise immune receptors outside their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment, induction of cytokine storms and T-cell anergy or apoptosis among many other effects. B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review of the structural basis for recognition of immune receptors by superantigens, we show that their binding interfaces share specific physicochemical characteristics when compared with other protein-protein interaction complexes. Given that antibody-binding superantigens have been exploited extensively in industrial antibody purification, these observations could facilitate further protein engineering to optimize the use of superantigens in this and other areas of biotechnology.
Collapse
Affiliation(s)
- Anthony M. Deacy
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Samuel Ken-En Gan
- Antibody & Product Development Lab, Experimental Drug Development Centre – Bioinformatics Institute (EDDC-BII), Agency for Science Technology and Research (ASTAR), Singapore, Singapore
- James Cook University, Singapore, Singapore
| | - Jeremy P. Derrick
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
15
|
Li F, Zhu M, Niu B, Liu L, Peng X, Yang H, Qin B, Wang M, Ren X, Zhou X. Generation and expression analysis of BAC humanized mice carrying HLA-DP401 haplotype. Animal Model Exp Med 2021; 4:116-128. [PMID: 34179719 PMCID: PMC8212823 DOI: 10.1002/ame2.12158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/17/2020] [Indexed: 11/13/2022] Open
Abstract
Background Human leukocyte antigen (HLA)-DP is much less studied than other HLA class II antigens, that is, HLA-DR and HLA-DQ, etc. However, the accumulating data have suggested the important roles of DP-restricted responses in the context of cancer, allergy, and infectious disease. Lack of animal models expressing these genes as authentic cis-haplotypes blocks our understanding for the role of HLA-DP haplotypes in immunity. Methods To explore the potential cis-acting control elements involved in the transcriptional regulation of the HLA-DPA1/DPB1 gene, we performed the expression analysis using bacterial artificial chromosome (BAC)-based transgenic humanized mice in the C57BL/6 background, which carried the entire HLA-DP401 gene locus. We further developed a mouse model of Staphylococcus aureus pneumonia in HLA-DP401 humanized transgenic mice, and performed the analysis on the expression pattern of HLA-DP401 and immunological responses in the model. Results In this study, we screened and identified a BAC clone spanning the entire HLA-DP gene locus. DNA from this clone was analyzed for integrity by pulsed-field gel electrophoresis and then microinjected into fertilized mouse oocytes to produce transgenic founder animals. Nine sets of PCR primers for regional markers with an average distance of 15 kb between each primer were used to confirm the integrity of the transgene in the five transgenic lines carrying the HLA-DPA1/DPB1 gene. Transgene copy numbers were determined by real-time PCR analysis. HLA-DP401 gene expression was analyzed at the mRNA and protein level. Although infection with S aureus Newman did not alter the percentage of immune cells in the spleen and thymus from the HLA-DP401-H2-Aβ1 humanized mice. Increased expression of HLA-DP401 was observed in the thymus of the humanized mice infected by S aureus. Conclusions We generated several BAC transgenic mice, and analyzed the expression of HLA-DPA1/DPB1 in those mice. A model of Saureus-induced pneumonia in the HLA-DP401-H2-Aβ1-/- humanized mice was further developed, and S aureus infection upregulated the HLA-DP401 expression in thymus of those humanized mice. These findings demonstrate the potential of those HLA-DPA1/DPB1 transgenic humanized mice for developing animal models of infectious diseases and MHC-associated immunological diseases.
Collapse
Affiliation(s)
- Feng Li
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Meng‐min Zhu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Bo‐wen Niu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Ling‐ling Liu
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Xiu‐hua Peng
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Hua Yang
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Bo‐yin Qin
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Meixiang Wang
- Department of Scientific ResearchShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Xiaonan Ren
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| | - Xiaohui Zhou
- Department of Laboratory Animal ScienceShanghai Public Health Clinical CenterFudan UniveristyShanghaiChina
| |
Collapse
|
16
|
The Phosphoarginine Phosphatase PtpB from Staphylococcus aureus Is Involved in Bacterial Stress Adaptation during Infection. Cells 2021; 10:cells10030645. [PMID: 33799337 PMCID: PMC8001253 DOI: 10.3390/cells10030645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus continues to be a public health threat, especially in hospital settings. Studies aimed at deciphering the molecular and cellular mechanisms that underlie pathogenesis, host adaptation, and virulence are required to develop effective treatment strategies. Numerous host-pathogen interactions were found to be dependent on phosphatases-mediated regulation. This study focused on the analysis of the role of the low-molecular weight phosphatase PtpB, in particular, during infection. Deletion of ptpB in S. aureus strain SA564 significantly reduced the capacity of the mutant to withstand intracellular killing by THP-1 macrophages. When injected into normoglycemic C57BL/6 mice, the SA564 ΔptpB mutant displayed markedly reduced bacterial loads in liver and kidney tissues in a murine S. aureus abscess model when compared to the wild type. We also observed that PtpB phosphatase-activity was sensitive to oxidative stress. Our quantitative transcript analyses revealed that PtpB affects the transcription of various genes involved in oxidative stress adaptation and infectivity. Thus, this study disclosed first insights into the physiological role of PtpB during host interaction allowing us to link phosphatase-dependent regulation to oxidative bacterial stress adaptation during infection.
Collapse
|
17
|
Population Analysis of Staphylococcus aureus Reveals a Cryptic, Highly Prevalent Superantigen SElW That Contributes to the Pathogenesis of Bacteremia. mBio 2020; 11:mBio.02082-20. [PMID: 33109757 PMCID: PMC7593966 DOI: 10.1128/mbio.02082-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Staphylococcus aureus is an important human and animal pathogen associated with an array of diseases, including life-threatening necrotizing pneumonia and infective endocarditis. The success of S. aureus as a pathogen has been linked in part to its ability to manipulate the host immune response through the secretion of toxins and immune evasion molecules. The staphylococcal superantigens (SAgs) have been studied for decades, but their role in S. aureus pathogenesis is not well understood, and an appreciation for how SAgs manipulate the host immune response to promote infection may be crucial for the development of novel intervention strategies. Here, we characterized a widely prevalent, previously cryptic, staphylococcal SAg, SElW, that contributes to the severity of S. aureus infections caused by an important epidemic clone of S. aureus CC398. Our findings add to the understanding of staphylococcal SAg diversity and function and provide new insights into the capacity of S. aureus to cause disease. Staphylococcal superantigens (SAgs) are a family of secreted toxins that stimulate T cell activation and are associated with an array of diseases in humans and livestock. Most SAgs produced by Staphylococcus aureus are encoded by mobile genetic elements, such as pathogenicity islands, bacteriophages, and plasmids, in a strain-dependent manner. Here, we carried out a population genomic analysis of >800 staphylococcal isolates representing the breadth of S. aureus diversity to investigate the distribution of all 26 identified SAg genes. Up to 14 SAg genes were identified per isolate with the most common gene selw (encoding a putative SAg, SElW) identified in 97% of isolates. Most isolates (62.5%) have a full-length open reading frame of selw with an alternative TTG start codon that may have precluded functional characterization of SElW to date. Here, we demonstrate that S. aureus uses the TTG start codon to translate a potent SAg SElW that induces Vβ-specific T cell proliferation, a defining feature of classical SAgs. SElW is the only SAg predicted to be expressed by isolates of the CC398 lineage, an important human and livestock epidemic clone. Deletion of selw in a representative CC398 clinical isolate, S. aureus NM001, resulted in complete loss of T cell mitogenicity in vitro, and in vivo expression of SElW by S. aureus increased the bacterial load in the liver during bloodstream infection of SAg-sensitive HLA-DR4 transgenic mice. Overall, we report the characterization of a novel, highly prevalent, and potent SAg that contributes to the pathogenesis of S. aureus infection.
Collapse
|
18
|
Mrochen DM, Fernandes de Oliveira LM, Raafat D, Holtfreter S. Staphylococcus aureus Host Tropism and Its Implications for Murine Infection Models. Int J Mol Sci 2020; 21:E7061. [PMID: 32992784 PMCID: PMC7582387 DOI: 10.3390/ijms21197061] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a pathobiont of humans as well as a multitude of animal species. The high prevalence of multi-resistant and more virulent strains of S. aureus necessitates the development of new prevention and treatment strategies for S. aureus infection. Major advances towards understanding the pathogenesis of S. aureus diseases have been made using conventional mouse models, i.e., by infecting naïve laboratory mice with human-adapted S.aureus strains. However, the failure to transfer certain results obtained in these murine systems to humans highlights the limitations of such models. Indeed, numerous S. aureus vaccine candidates showed promising results in conventional mouse models but failed to offer protection in human clinical trials. These limitations arise not only from the widely discussed physiological differences between mice and humans, but also from the lack of attention that is paid to the specific interactions of S. aureus with its respective host. For instance, animal-derived S. aureus lineages show a high degree of host tropism and carry a repertoire of host-specific virulence and immune evasion factors. Mouse-adapted S.aureus strains, humanized mice, and microbiome-optimized mice are promising approaches to overcome these limitations and could improve transferability of animal experiments to human trials in the future.
Collapse
Affiliation(s)
- Daniel M. Mrochen
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
| | - Liliane M. Fernandes de Oliveira
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
| | - Dina Raafat
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, 21521 Alexandria, Egypt
| | - Silva Holtfreter
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
| |
Collapse
|
19
|
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: 16] [Impact Index Per Article: 3.2] [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
|
20
|
Research Techniques Made Simple: Mouse Bacterial Skin Infection Models for Immunity Research. J Invest Dermatol 2020; 140:1488-1497.e1. [PMID: 32407714 DOI: 10.1016/j.jid.2020.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/11/2023]
Abstract
Bacterial skin infections are a major societal health burden and are increasingly difficult to treat owing to the emergence of antibiotic-resistant strains such as community-acquired methicillin-resistant Staphylococcus aureus. Understanding the immunologic mechanisms that provide durable protection against skin infections has the potential to guide the development of immunotherapies and vaccines to engage the host immune response to combat these antibiotic-resistant strains. To this end, mouse skin infection models allow researchers to examine host immunity by investigating the timing, inoculum, route of infection and the causative bacterial species in different wild-type mouse backgrounds as well as in knockout, transgenic, and other types of genetically engineered mouse strains. To recapitulate the various types of human skin infections, many different mouse models have been developed. For example, four models frequently used in dermatological research are based on the route of infection, including (i) subcutaneous infection models, (ii) intradermal infection models, (iii) wound infection models, and (iv) epicutaneous infection models. In this article, we will describe these skin infection models in detail along with their advantages and limitations. In addition, we will discuss how humanized mouse models such as the human skin xenograft on immunocompromised mice might be used in bacterial skin infection research.
Collapse
|
21
|
Monistero V, Barberio A, Biscarini F, Cremonesi P, Castiglioni B, Graber HU, Bottini E, Ceballos-Marquez A, Kroemker V, Petzer IM, Pollera C, Santisteban C, Veiga Dos Santos M, Bronzo V, Piccinini R, Re G, Cocchi M, Moroni P. Different distribution of antimicrobial resistance genes and virulence profiles of Staphylococcus aureus strains isolated from clinical mastitis in six countries. J Dairy Sci 2020; 103:3431-3446. [PMID: 32008788 DOI: 10.3168/jds.2019-17141] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 12/03/2019] [Indexed: 11/19/2022]
Abstract
Staphylococcus aureus is recognized worldwide as one of the main contagious mastitis agents in cattle and can express a set of antimicrobial resistance genes and virulence-associated genes that explain the wide range of outcomes of intramammary infections. Staphylococcus aureus strains are heterogeneous: their different resistance and virulence patterns, associated with host-level factors and treatment factors, are related to the severity of infection. The aim of this study was to determine phenotypic antibiotic susceptibility, occurrence of selected antimicrobial resistance genes and other virulence genes in 93 S. aureus strains isolated from clinical mastitis in 6 countries: Argentina, Brazil, Germany, Italy, the United States (New York State), and South Africa. These isolates were tested against a total of 16 drugs (amoxicillin-clavulanate, ampicillin, cefazolin, cefoperazone, cefquinome, enrofloxacin, erythromycin, gentamicin, kanamycin, lincomycin, oxacillin, penicillin, rifampin, spiramycin, sulfamethoxazole/trimethoprim, tylosin) by minimum inhibitory concentration (MIC) assay, and examined for the presence of 6 antibiotic-resistance genes (blaZ, mecA, mecC, ermA, ermB, ermC) and 6 virulence-associated genes (scn, chp, sak, hla, hlb, sea) via PCR analysis. The phenotypic results of this study revealed the presence of 19.4% penicillin-resistant strains, whereas 22.6% of the strains were classified as having resistance (5.4%) or intermediate resistance (17.2%) to erythromycin. Most (96.8%) of the isolates were inhibited by cephalosporins, and all were susceptible to amoxicillin-clavulanate. Two strains (1 from Germany, 1 from Italy) were resistant to oxacillin and were positive for mecA. Among the other antimicrobial resistance genes, the most frequently detected was blaZ (46.2%), and 32.3% of the isolates were positive for erm genes: ermC (21.5%) and ermB (10.8%). The most prevalent virulence gene was hla (100%), followed by hlb (84.9%) and sea (65.6%). These results show a low prevalence of antibiotic multidrug resistance in S. aureus isolates, even if the detection of selected antimicrobial resistance genes did not always correspond with the occurrence of phenotypic antibiotic resistance; the immune evasion cluster gene prevalence was quite low in the samples analyzed.
Collapse
Affiliation(s)
- V Monistero
- Università degli Studi di Milano, Dipartimento di Medicina Veterinaria, Italy, 20133
| | - A Barberio
- Istituto Zooprofilattico Sperimentale delle Venezie, Sezione Territoriale di Padova, Legnaro (PD), Italy, 3020
| | - F Biscarini
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy, 26900
| | - P Cremonesi
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy, 26900
| | - B Castiglioni
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy, 26900
| | - H U Graber
- Agroscope, Research Division, Food Microbial Systems, Bern, Switzerland, 3003
| | - E Bottini
- Laboratorio de Microbiologia Clinica y Experimental, Departamento de Sanidad Animal y Medicina Preventiva SAMP/CIVENTAN, Becaria CONICET, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (FCV, UNCPBA), Paraje Arroyo Seco S/N, Campus Universitario, CP 7000 Tandil, Argentina
| | - A Ceballos-Marquez
- Laboratorio de Calidad de Leche y Epidemiología Veterinaria (Grupo CLEV), Universidad de 6 Caldas, Manizales, 170003, Colombia
| | - V Kroemker
- Bioprocess Engineering-Faculty II, Microbiology, University of Applied Sciences and Arts, 30453 Hannover, Germany
| | - I M Petzer
- Faculty of Veterinary Science, Onderstepoort, University of Pretoria, 0110, South Africa
| | - C Pollera
- Università degli Studi di Milano, Dipartimento di Medicina Veterinaria, Italy, 20133
| | - C Santisteban
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, NY 14850
| | - M Veiga Dos Santos
- Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Sciences, Pirassununga-SP 13635900, Brazil
| | - V Bronzo
- Università degli Studi di Milano, Dipartimento di Medicina Veterinaria, Italy, 20133
| | - R Piccinini
- Università degli Studi di Milano, Dipartimento di Medicina Veterinaria, Italy, 20133
| | - G Re
- Università degli Studi di Torino, Dipartimento di Scienze Veterinarie, Turin, Italy, 10085
| | - M Cocchi
- Istituto Zooprofilattico Sperimentale delle Venezie, Sezione Territoriale di Udine, Basaldella di Campoformido (UD), Italy, 33030
| | - P Moroni
- Università degli Studi di Milano, Dipartimento di Medicina Veterinaria, Italy, 20133; Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, NY 14850.
| |
Collapse
|
22
|
Meilleur CE, Wardell CM, Mele TS, Dikeakos JD, Bennink JR, Mu HH, McCormick JK, Haeryfar SMM. Bacterial Superantigens Expand and Activate, Rather than Delete or Incapacitate, Preexisting Antigen-Specific Memory CD8+ T Cells. J Infect Dis 2020; 219:1307-1317. [PMID: 30418594 DOI: 10.1093/infdis/jiy647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/07/2018] [Indexed: 11/13/2022] Open
Abstract
Superantigens (SAgs) released by common Gram-positive bacterial pathogens have been reported to delete, anergize, or activate mouse T cells. However, little is known about their effects on preexisting memory CD8+ T cell (TCD8) pools. Furthermore, whether SAgs manipulate human memory TCD8 responses to cognate antigens is unknown. We used a human peripheral blood mononuclear cell culture system and a nontransgenic mouse model in which the impact of stimulation by two fundamentally distinct SAgs, staphylococcal enterotoxin B and Mycoplasma arthritidis mitogen, on influenza virus- and/or cytomegalovirus-specific memory TCD8 could be monitored. Bacterial SAgs surprisingly expanded antiviral memory TCD8 generated naturally through infection or artificially through vaccination. Mechanistically, this was a T cell-intrinsic and T cell receptor β-chain variable-dependent phenomenon. Importantly, SAg-expanded TCD8 displayed an effector memory phenotype and were capable of producing interferon-γ and destroying target cells ex vivo or in vivo. These findings have clear implications for antimicrobial defense and rational vaccine design.
Collapse
Affiliation(s)
- Courtney E Meilleur
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Christine M Wardell
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Tina S Mele
- Division of General Surgery, Department of Surgery, Western University, London, Canada.,Division of Critical Care Medicine, Western University, London, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Jack R Bennink
- Viral Immunology Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Hong-Hua Mu
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City
| | - John K McCormick
- Department of Microbiology and Immunology, Western University, London, Canada.,Centre for Human Immunology, Western University, London, Canada.,Lawson Health Research Institute, London, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Canada.,Division of General Surgery, Department of Surgery, Western University, London, Canada.,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Canada.,Centre for Human Immunology, Western University, London, Canada.,Lawson Health Research Institute, London, Canada
| |
Collapse
|
23
|
Toxic Shock Syndrome Toxin 1 Evaluation and Antibiotic Impact in a Transgenic Model of Staphylococcal Soft Tissue Infection. mSphere 2019; 4:4/5/e00665-19. [PMID: 31597722 PMCID: PMC6796978 DOI: 10.1128/msphere.00665-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Staphylococcal toxic shock syndrome (TSS) is a life-threatening illness causing fever, rash, and shock, attributed to toxins produced by the bacterium Staphylococcus aureus, mainly toxic shock syndrome toxin 1 (TSST-1). TSS was in the past commonly linked with menstruation and high-absorbency tampons; now, TSS is more frequently triggered by other staphylococcal infections, particularly of skin and soft tissue. Investigating the progress and treatment of TSS in patients is challenging, as TSS is rare; animal models do not mimic TSS adequately, as toxins interact best with human immune cells. We developed a new model of staphylococcal soft tissue infection in mice producing human immune cell proteins, rendering them TSST-1 sensitive, to investigate TSS. The significance of our research was that TSST-1 was found in soft tissues and immune organs of mice and that early treatment of mice with the antibiotic clindamycin altered TSST-1 production. Therefore, the early treatment of patients suspected of having TSS with clindamycin may influence their response to treatment. Nonmenstrual toxic shock syndrome (nmTSS), linked to TSST-1-producing CC30 Staphylococcus aureus, is the leading manifestation of toxic shock syndrome (TSS). Due to case rarity and a lack of tractable animal models, TSS pathogenesis is poorly understood. We developed an S. aureus abscess model in HLA class II transgenic mice to investigate pathogenesis and treatment. TSST-1 sensitivity was established using murine spleen cell proliferation assays and cytokine assays following TSST-1 injection in vivo. HLA-DQ8 mice were infected subcutaneously with a tst-positive CC30 methicillin-sensitive S. aureus clinical TSS-associated isolate. Mice received intraperitoneal flucloxacillin, clindamycin, flucloxacillin and clindamycin, or a control reagent. Abscess size, bacterial counts, TSST-1 expression, and TSST-1 bioactivity were measured in tissues. Antibiotic effects were compared with the effects of control reagent. Purified TSST-1 expanded HLA-DQ8 T-cell Vβ subsets 3 and 13 in vitro and instigated cytokine release in vivo, confirming TSST-1 sensitivity. TSST-1 was detected in abscesses (0 to 8.0 μg/ml) and draining lymph nodes (0 to 0.2 μg/ml) of infected mice. Interleukin 6 (IL-6), gamma interferon (IFN-γ), KC (CXCL1), and MCP-1 were consistent markers of inflammation during infection. Clindamycin-containing antibiotic regimens reduced abscess size and TSST-1 production. Infection led to detectable TSST-1 in soft tissues, and TSST-1 was detected in draining lymph nodes, events which may be pivotal to TSS pathogenesis. The reduction in TSST-1 production and lesion size after a single dose of clindamycin underscores a potential role for adjunctive clindamycin at the start of treatment of patients suspected of having TSS to alter disease progression. IMPORTANCE Staphylococcal toxic shock syndrome (TSS) is a life-threatening illness causing fever, rash, and shock, attributed to toxins produced by the bacterium Staphylococcus aureus, mainly toxic shock syndrome toxin 1 (TSST-1). TSS was in the past commonly linked with menstruation and high-absorbency tampons; now, TSS is more frequently triggered by other staphylococcal infections, particularly of skin and soft tissue. Investigating the progress and treatment of TSS in patients is challenging, as TSS is rare; animal models do not mimic TSS adequately, as toxins interact best with human immune cells. We developed a new model of staphylococcal soft tissue infection in mice producing human immune cell proteins, rendering them TSST-1 sensitive, to investigate TSS. The significance of our research was that TSST-1 was found in soft tissues and immune organs of mice and that early treatment of mice with the antibiotic clindamycin altered TSST-1 production. Therefore, the early treatment of patients suspected of having TSS with clindamycin may influence their response to treatment.
Collapse
|
24
|
Anjum MF, Marco-Jimenez F, Duncan D, Marín C, Smith RP, Evans SJ. Livestock-Associated Methicillin-Resistant Staphylococcus aureus From Animals and Animal Products in the UK. Front Microbiol 2019; 10:2136. [PMID: 31572341 PMCID: PMC6751287 DOI: 10.3389/fmicb.2019.02136] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 08/30/2019] [Indexed: 01/08/2023] Open
Abstract
Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) is an emerging problem in many parts of the world. Although animal-adapted LA-MRSA has been known for many years, recent reports suggest a possible increasing trend in the zoonotic transmission of LA-MRSA in Europe. Since its emergence in the early 2000's, several investigations have indicated that persons in prolonged, repeated contact with affected livestock are at a higher risk of becoming colonized with LA-MRSA. LA-MRSA monitoring in livestock is voluntary under current EU legislation, and not all member states, including the UK, participate. UK LA-MRSA isolates have been detected through scanning surveillance, where samples are submitted from clinically diseased livestock for diagnostic investigation, and research studies. Surveys conducted on retail beef, pig and poultry meat on sale in the UK have also detected LA-MRSA. Taken together these results suggest that LA-MRSA is present in the UK, possibly at low prevalence level, as suggested by available evidence. In this review, we examine the data available from UK livestock and animal products, and make recommendations for future. We also review the findings from whole genome sequencing (WGS) of the possible lineage of some UK livestock isolates.
Collapse
Affiliation(s)
- Muna F. Anjum
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Francisco Marco-Jimenez
- Instituto de Ciencia y Tecnología Animal, Universitat Politècnica de València, Valencia, Spain
- Department of Epidemiological Sciences, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Daisy Duncan
- Department of Epidemiological Sciences, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Clara Marín
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
- Departamento de Producción Animal, Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Instituto de Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Richard P. Smith
- Department of Epidemiological Sciences, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Sarah J. Evans
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| |
Collapse
|
25
|
Radcliff FJ, Waldvogel-Thurlow S, Clow F, Mahadevan M, Johnston J, Li G, Proft T, Douglas RG, Fraser JD. Impact of Superantigen-Producing Bacteria on T Cells from Tonsillar Hyperplasia. Pathogens 2019; 8:pathogens8030090. [PMID: 31252586 PMCID: PMC6789895 DOI: 10.3390/pathogens8030090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus and Group A Streptococcus (GAS) are common occupants of the tonsils and many strains produce potent exotoxins (mitogens) that directly target T cells, which could be a driver for tonsillar hyperplasia. Tonsil tissues from 41 patients were tested for these bacteria in conjunction with profiling of B and T cells by flow cytometry. S. aureus and GAS were detected in tonsil tissue from 44% and 7%, respectively, of patients by bacteriological culture; immuno-histology showed bacteria in close proximity to both B and T lymphocytes. The presence of tonsillar S. aureus did not alter B or T cell populations, whereas peripheral blood mucosal-associated invariant T (MAIT) cells were significantly increased in S. aureus culture positive individuals (p < 0.006). Alterations of tonsil CD4+ TCR Vβ family members relative to peripheral blood were evident in 29 patients. Three patients had strong TCR Vβ skewing indicative of recent exposure to superantigens, their tonsils contained mitogenic bacteria, and supernatants from these bacteria were used to partially recapitulate the skewing profile in vitro, supporting the notion that superantigens can target tonsillar T cells in situ. Tonsils are a reservoir for superantigen-producing bacteria with the capacity to alter the composition and function of key immune cells.
Collapse
Affiliation(s)
- Fiona J Radcliff
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand.
| | | | - Fiona Clow
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Murali Mahadevan
- Department of Surgery, University of Auckland, Auckland 1023, New Zealand
| | - James Johnston
- Department of Surgery, University of Auckland, Auckland 1023, New Zealand
| | - Gen Li
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Richard G Douglas
- Department of Surgery, University of Auckland, Auckland 1023, New Zealand
| | - John D Fraser
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
26
|
Staphylococcal Superantigens: Pyrogenic Toxins Induce Toxic Shock. Toxins (Basel) 2019; 11:toxins11030178. [PMID: 30909619 PMCID: PMC6468478 DOI: 10.3390/toxins11030178] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 01/01/2023] Open
Abstract
Staphylococcal enterotoxin B (SEB) and related superantigenic toxins produced by Staphylococcus aureus are potent activators of the immune system. These protein toxins bind to major histocompatibility complex (MHC) class II molecules and specific Vβ regions of T-cell receptors (TCRs), resulting in the activation of both monocytes/macrophages and T lymphocytes. The bridging of TCRs with MHC class II molecules by superantigens triggers an early “cytokine storm” and massive polyclonal T-cell proliferation. Proinflammatory cytokines, tumor necrosis factor α, interleukin 1 (IL-1), IL-2, interferon γ (IFNγ), and macrophage chemoattractant protein 1 elicit fever, inflammation, multiple organ injury, hypotension, and lethal shock. Upon MHC/TCR ligation, superantigens induce signaling pathways, including mitogen-activated protein kinase cascades and cytokine receptor signaling, which results in NFκB activation and the phosphoinositide 3-kinase/mammalian target of rapamycin pathways. In addition, gene profiling studies have revealed the essential roles of innate antimicrobial defense genes in the pathogenesis of SEB. The genes expressed in a murine model of SEB-induced shock include intracellular DNA/RNA sensors, apoptosis/DNA damage-related molecules, endoplasmic reticulum/mitochondrial stress responses, immunoproteasome components, and IFN-stimulated genes. This review focuses on the signaling pathways induced by superantigens that lead to the activation of inflammation and damage response genes. The induction of these damage response genes provides evidence that SEB induces danger signals in host cells, resulting in multiorgan injury and toxic shock. Therapeutics targeting both host inflammatory and cell death pathways can potentially mitigate the toxic effects of staphylococcal superantigens.
Collapse
|
27
|
Paik W, Alonzo F, Knight KL. Probiotic Exopolysaccharide Protects against Systemic Staphylococcus aureus Infection, Inducing Dual-Functioning Macrophages That Restrict Bacterial Growth and Limit Inflammation. Infect Immun 2019; 87:e00791-18. [PMID: 30396894 PMCID: PMC6300633 DOI: 10.1128/iai.00791-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 10/26/2018] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus causes severe systemic infection with high mortality rates. We previously identified exopolysaccharide (EPS) from a probiotic, Bacillus subtilis, that induces anti-inflammatory macrophages with an M2 phenotype and protects mice from Citrobacter rodentium-induced colitis. We tested if EPS could protect from systemic infection induced by S. aureus and found that EPS-treated mice had enhanced survival as well as reduced weight loss, systemic inflammation, and bacterial burden. While macrophages from EPS-treated mice display an M2 phenotype, they also restrict growth of internalized S. aureus through reactive oxygen species (ROS), reminiscent of proinflammatory phagocytes. These EPS-induced macrophages also limit T cell activation by S. aureus superantigens, and EPS abrogates systemic induction of gamma interferon after infection. We conclude that B. subtilis EPS is an immunomodulatory agent that induces hybrid macrophages that bolster antibacterial immunity and simultaneously limit inflammation, reducing disease burden and promoting host survival.
Collapse
Affiliation(s)
- Wonbeom Paik
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Francis Alonzo
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| |
Collapse
|
28
|
An TJ, Benvenuti MA, Mignemi ME, Thomsen IP, Schoenecker JG. Pediatric Musculoskeletal Infection: Hijacking the Acute-Phase Response. JBJS Rev 2018; 4:01874474-201609000-00004. [PMID: 27760072 DOI: 10.2106/jbjs.rvw.15.00099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tissue injury activates the acute-phase response mediated by the liver, which promotes coagulation, immunity, and tissue regeneration. To survive and disseminate, musculoskeletal pathogens express virulence factors that modulate and hijack this response. As the acute-phase reactants required by these pathogens are most abundant in damaged tissue, these infections are predisposed to occur in tissues following traumatic or surgical injury. Staphylococcus aureus expresses the virulence factors coagulase and von Willebrand binding protein to stimulate coagulation and to form a fibrin abscess that protects it from host immune-cell phagocytosis. After the staphylococcal abscess community reaches quorum, which is the colony density that enables cell-to-cell communication and coordinated gene expression, subsequent expression of staphylokinase stimulates activation of fibrinolysis, which ruptures the abscess wall and results in bacterial dissemination. Unlike Staphylococcus aureus, Streptococcus pyogenes expresses streptokinase and other virulence factors to activate fibrinolysis and to rapidly disseminate throughout the body, causing diseases such as necrotizing fasciitis. Understanding the virulence strategies of musculoskeletal pathogens will help to guide clinical diagnosis and decision-making through monitoring of acute-phase markers such as C-reactive protein, erythrocyte sedimentation rate, and fibrinogen.
Collapse
Affiliation(s)
- Thomas J An
- Departments of Orthopaedics (M.E.M. and J.G.S.), Pediatrics (I.P.T. and J.G.S.), Pediatric Infectious Disease (I.P.T.), Pharmacology (J.G.S.), and Pathology (J.G.S.), Vanderbilt University School of Medicine (T.J.A. and M.A.B.), Nashville, Tennessee
| | | | | | | | | |
Collapse
|
29
|
Surewaard BGJ, Thanabalasuriar A, Zeng Z, Tkaczyk C, Cohen TS, Bardoel BW, Jorch SK, Deppermann C, Bubeck Wardenburg J, Davis RP, Jenne CN, Stover KC, Sellman BR, Kubes P. α-Toxin Induces Platelet Aggregation and Liver Injury during Staphylococcus aureus Sepsis. Cell Host Microbe 2018; 24:271-284.e3. [PMID: 30033122 DOI: 10.1016/j.chom.2018.06.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/15/2018] [Accepted: 06/27/2018] [Indexed: 01/09/2023]
Abstract
During sepsis, small blood vessels can become occluded by large platelet aggregates of poorly understood etiology. During Staphylococcal aureus infection, sepsis severity is linked to the bacterial α-toxin (α-hemolysin, AT) through unclear mechanisms. In this study, we visualized intravascular events in the microcirculation and found that intravenous AT injection induces rapid platelet aggregation, forming dynamic micro-thrombi in the microcirculation. These aggregates are retained in the liver sinusoids and kidney glomeruli, causing multi-organ dysfunction. Acute staphylococcal infection results in sequestration of most bacteria by liver macrophages. Platelets are initially recruited to these macrophages and help eradicate S. aureus. However, at later time points, AT causes aberrant and damaging thrombosis throughout the liver. Treatment with an AT neutralizing antibody (MEDI4893∗) prevents platelet aggregation and subsequent liver damage, without affecting the initial and beneficial platelet recruitment. Thus, AT neutralization may represent a promising approach to combat staphylococcal-induced intravascular coagulation and organ dysfunction.
Collapse
Affiliation(s)
- Bas G J Surewaard
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Medical Microbiology, University Medical Center, Utrecht, the Netherlands
| | - Ajitha Thanabalasuriar
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Zhutian Zeng
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Christine Tkaczyk
- Department of Microbial Sciences, Medimmune, LLC, Gaithersburg, MD, USA
| | - Taylor S Cohen
- Department of Microbial Sciences, Medimmune, LLC, Gaithersburg, MD, USA
| | - Bart W Bardoel
- Department of Medical Microbiology, University Medical Center, Utrecht, the Netherlands
| | - Selina K Jorch
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Carsten Deppermann
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Juliane Bubeck Wardenburg
- Department of Pediatrics and Division of Pediatric Critical Care Medicine at Washington University School of Medicine, St. Louis, MO, USA
| | - Rachelle P Davis
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, AB, Canada
| | - Craig N Jenne
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, AB, Canada
| | - Kendall C Stover
- Department of Microbial Sciences, Medimmune, LLC, Gaithersburg, MD, USA
| | - Bret R Sellman
- Department of Microbial Sciences, Medimmune, LLC, Gaithersburg, MD, USA
| | - Paul Kubes
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
30
|
Tuffs SW, Haeryfar SMM, McCormick JK. Manipulation of Innate and Adaptive Immunity by Staphylococcal Superantigens. Pathogens 2018; 7:pathogens7020053. [PMID: 29843476 PMCID: PMC6027230 DOI: 10.3390/pathogens7020053] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Staphylococcal superantigens (SAgs) constitute a family of potent exotoxins secreted by Staphylococcus aureus and other select staphylococcal species. SAgs function to cross-link major histocompatibility complex (MHC) class II molecules with T cell receptors (TCRs) to stimulate the uncontrolled activation of T lymphocytes, potentially leading to severe human illnesses such as toxic shock syndrome. The ubiquity of SAgs in clinical S. aureus isolates suggests that they likely make an important contribution to the evolutionary fitness of S. aureus. Although the apparent redundancy of SAgs in S. aureus has not been explained, the high level of sequence diversity within this toxin family may allow for SAgs to recognize an assorted range of TCR and MHC class II molecules, as well as aid in the avoidance of humoral immunity. Herein, we outline the major diseases associated with the staphylococcal SAgs and how a dysregulated immune system may contribute to pathology. We then highlight recent research that considers the importance of SAgs in the pathogenesis of S. aureus infections, demonstrating that SAgs are more than simply an immunological diversion. We suggest that SAgs can act as targeted modulators that drive the immune response away from an effective response, and thus aid in S. aureus persistence.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON N6A 3K7, Canada.
- Centre for Human Immunology, Western University, London, ON N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON N6C 2R5, Canada.
| | - John K McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, Western University, London, ON N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON N6C 2R5, Canada.
| |
Collapse
|
31
|
Ferreira-Duarte AP, Pinheiro-Torres AS, Anhê GF, Condino-Neto A, Antunes E, DeSouza IA. MHC Class II Activation and Interferon-γ Mediate the Inhibition of Neutrophils and Eosinophils by Staphylococcal Enterotoxin Type A (SEA). Front Cell Infect Microbiol 2017; 7:518. [PMID: 29322036 PMCID: PMC5733477 DOI: 10.3389/fcimb.2017.00518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/04/2017] [Indexed: 12/30/2022] Open
Abstract
Staphylococcal enterotoxins are classified as superantigens that act by linking T-cell receptor with MHC class II molecules, which are expressed on classical antigen-presenting cells (APC). Evidence shows that MHC class II is also expressed in neutrophils and eosinophils. This study aimed to investigate the role of MHC class II and IFN-γ on chemotactic and adhesion properties of neutrophils and eosinophils after incubation with SEA. Bone marrow (BM) cells obtained from BALB/c mice were resuspended in culture medium, and incubated with SEA (3–30 ng/ml; 1–4 h), after which chemotaxis and adhesion were evaluated. Incubation with SEA significantly reduced the chemotactic and adhesive responses in BM neutrophils activated with IL-8 (200 ng/ml). Likewise, SEA significantly reduced the chemotactic and adhesive responses of BM eosinophils activated with eotaxin (300 ng/ml). The inhibitory effects of SEA on cell chemotaxis and adhesion were fully prevented by prior incubation with an anti-MHC class II blocking antibody (2 μg/ml). SEA also significantly reduced the intracellular Ca2+ levels in IL-8- and eotaxin-activated BM cells. No alterations of MAC-1, VLA4, and LFA-1α expressions were observed after SEA incubation. In addition, SEA elevated by 3.5-fold (P < 0.05) the INF-γ levels in BM cells. Incubation of BM leukocytes with IFN-γ (10 ng/ml, 2 h) reduced both neutrophil and eosinophil chemotaxis and adhesion, which were prevented by prior incubation with anti-MHC class II antibody (2 μg/ml). In conclusion, SEA inhibits neutrophil and eosinophil by MHC class II-dependent mechanism, which may be modulated by concomitant release of IFN-γ.
Collapse
Affiliation(s)
- Ana P Ferreira-Duarte
- Department of Biology and Physiology, Faculty of Medicine of Jundiai, Jundiai, Brazil
| | | | - Gabriel F Anhê
- Department of Pharmacology, State University of Campinas, Campinas, Brazil
| | - Antônio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, State University of Campinas, Campinas, Brazil
| | - Ivani A DeSouza
- Department of Biology and Physiology, Faculty of Medicine of Jundiai, Jundiai, Brazil
| |
Collapse
|
32
|
Langley RJ, Ting YT, Clow F, Young PG, Radcliff FJ, Choi JM, Sequeira RP, Holtfreter S, Baker H, Fraser JD. Staphylococcal enterotoxin-like X (SElX) is a unique superantigen with functional features of two major families of staphylococcal virulence factors. PLoS Pathog 2017; 13:e1006549. [PMID: 28880913 PMCID: PMC5589262 DOI: 10.1371/journal.ppat.1006549] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 07/24/2017] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that produces many virulence factors. Two major families of which are the staphylococcal superantigens (SAgs) and the Staphylococcal Superantigen-Like (SSL) exoproteins. The former are immunomodulatory toxins that induce a Vβ-specific activation of T cells, while the latter are immune evasion molecules that interfere with a wide range of innate immune defences. The superantigenic properties of Staphylococcal enterotoxin-like X (SElX) have recently been established. We now reveal that SElX also possesses functional characteristics of the SSLs. A region of SElX displays high homology to the sialyl-lactosamine (sLacNac)-specific binding site present in a sub-family of SSLs. By analysing the interaction of SElX with sLacNac-containing glycans we show that SElX has an equivalent specificity and host cell binding range to the SSLs. Mutation of key amino acids in this conserved region affects the ability of SElX to bind to cells of myeloid origin and significantly reduces its ability to protect S. aureus from destruction in a whole blood killing (WBK) assay. Like the SSLs, SElX is up-regulated early during infection and is under the control of the S. aureus exotoxin expression (Sae) two component gene regulatory system. Additionally, the structure of SElX in complex with the sLacNac-containing tetrasaccharide sialyl Lewis X (sLeX) reveals that SElX is a unique single-domain SAg. In summary, SElX is an ‘SSL-like’ SAg. The ability of Staphylococcus aureus to cause disease can be attributed to the wide range of toxins and immune evasion molecules it produces. The 25-member superantigen (SAg) family of toxins disrupts adaptive immunity by activating large proportions of T cells. In contrast, the structurally-related 14-member Staphylococcal Superantigen-Like (SSL) family inhibits a wide range of innate immune functions. We have discovered that the SAg staphylococcal enterotoxin-like X (SElX) has the sialylated-glycan-dependent active site found in a sub-family of SSLs. Through this site it possesses the ability to affect host innate immunity defences. By solving the X-ray crystal structure of SElX we have also discovered that SElX is a unique single-domain SAg. While it retains a typical β-grasp domain, it lacks the OB-fold domain that is present in all other staphylococcal SAgs.
Collapse
Affiliation(s)
- Ries J. Langley
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
- * E-mail:
| | - Yi Tian Ting
- School of Biological Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Fiona Clow
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Paul G. Young
- School of Biological Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Fiona J. Radcliff
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Jeong Min Choi
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Richard P. Sequeira
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Silva Holtfreter
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Heather Baker
- School of Biological Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - John D. Fraser
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
33
|
Tuffs SW, James DBA, Bestebroer J, Richards AC, Goncheva MI, O’Shea M, Wee BA, Seo KS, Schlievert PM, Lengeling A, van Strijp JA, Torres VJ, Fitzgerald JR. The Staphylococcus aureus superantigen SElX is a bifunctional toxin that inhibits neutrophil function. PLoS Pathog 2017; 13:e1006461. [PMID: 28880920 PMCID: PMC5589267 DOI: 10.1371/journal.ppat.1006461] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/09/2017] [Indexed: 12/29/2022] Open
Abstract
Bacterial superantigens (SAgs) cause Vβ-dependent T-cell proliferation leading to immune dysregulation associated with the pathogenesis of life-threatening infections such as toxic shock syndrome, and necrotizing pneumonia. Previously, we demonstrated that staphylococcal enterotoxin-like toxin X (SElX) from Staphylococcus aureus is a classical superantigen that exhibits T-cell activation in a Vβ-specific manner, and contributes to the pathogenesis of necrotizing pneumonia. Here, we discovered that SElX can also bind to neutrophils from human and other mammalian species and disrupt IgG-mediated phagocytosis. Site-directed mutagenesis of the conserved sialic acid-binding motif of SElX abolished neutrophil binding and phagocytic killing, and revealed multiple glycosylated neutrophil receptors for SElX binding. Furthermore, the neutrophil binding-deficient mutant of SElX retained its capacity for T-cell activation demonstrating that SElX exhibits mechanistically independent activities on distinct cell populations associated with acquired and innate immunity, respectively. Finally, we demonstrated that the neutrophil-binding activity rather than superantigenicity is responsible for the SElX-dependent virulence observed in a necrotizing pneumonia rabbit model of infection. Taken together, we report the first example of a SAg, that can manipulate both the innate and adaptive arms of the human immune system during S. aureus pathogenesis. Staphylococcus aureus is a bacterial pathogen responsible for an array of disease types in healthcare and community settings. One of the keys to the success of this pathogen is its ability to subvert the immune system of the host. Here we demonstrate that the superantigen (SAg) staphylococcal enterotoxin-like toxin X (SElX) contributes to immune evasion by inducing unregulated T-cell proliferation, and by inhibition of phagocytosis by neutrophils. We observed that the capacity to bind neutrophils appears to be central to the SElX-dependent toxicity observed in a necrotising pneumonia infection model in rabbits. We report the first example of a staphylococcal SAg with two independent immunomodulatory functions acting on distinct immune cell types.
Collapse
Affiliation(s)
- Stephen W. Tuffs
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
| | - David B. A. James
- Department of Microbiology, New York University School of Medicine, New York, NY, United Kingdom
| | - Jovanka Bestebroer
- Department Medical Microbiology, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Amy C. Richards
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
| | - Mariya I. Goncheva
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
| | - Marie O’Shea
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
| | - Bryan A. Wee
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
| | - Keun Seok Seo
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Patrick M. Schlievert
- Department of Microbiology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Andreas Lengeling
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
| | - Jos A. van Strijp
- Department Medical Microbiology, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, New York, NY, United Kingdom
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, Scotland, United States of America
- * E-mail:
| |
Collapse
|
34
|
Sandberg JK, Norrby-Teglund A, Leeansyah E. Bacterial deception of MAIT cells in a cloud of superantigen and cytokines. PLoS Biol 2017; 15:e2003167. [PMID: 28742082 PMCID: PMC5542701 DOI: 10.1371/journal.pbio.2003167] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/03/2017] [Indexed: 01/06/2023] Open
Abstract
The bacterium Staphylococcus aureus is an important cause of the life-threatening condition toxic shock syndrome in humans. Bacterial toxins known as superantigens (SAgs) generate this illness by acting as broad activators of a substantial fraction of all T lymphocytes, bypassing the normally highly stringent T-cell receptor antigen specificity to cause a systemic inflammatory cytokine storm in the host. In a new study, Shaler et al. found that immune cells called mucosa-associated invariant T (MAIT) cells make an unexpectedly large contribution to the SAg response in a largely T-cell receptor–independent, cytokine-driven manner. Subsequent to such activation, the MAIT cells remain unresponsive to stimulation with bacterial antigen. Thus, S. aureus hijacks MAIT cells in the cytokine storm and leaves them functionally impaired. This work provides new insight into the role of MAIT cells in antibacterial immunity and opens new avenues of investigation to understand and possibly treat bacterial toxic shock and sepsis.
Collapse
Affiliation(s)
- Johan K. Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- * E-mail:
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Edwin Leeansyah
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| |
Collapse
|
35
|
Suliman HB, Kraft B, Bartz R, Chen L, Welty-Wolf KE, Piantadosi CA. Mitochondrial quality control in alveolar epithelial cells damaged by S. aureus pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2017; 313:L699-L709. [PMID: 28663335 DOI: 10.1152/ajplung.00197.2017] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/30/2022] Open
Abstract
Mitochondrial damage is often overlooked in acute lung injury (ALI), yet most of the lung's physiological processes, such as airway tone, mucociliary clearance, ventilation-perfusion (Va/Q) matching, and immune surveillance require aerobic energy provision. Because the cell's mitochondrial quality control (QC) process regulates the elimination and replacement of damaged mitochondria to maintain cell survival, we serially evaluated mitochondrial biogenesis and mitophagy in the alveolar regions of mice in a validated Staphylococcus aureus pneumonia model. We report that apart from cell lysis by direct contact with microbes, modest epithelial cell death was detected despite significant mitochondrial damage. Cell death by TdT-mediated dUTP nick-end labeling staining occurred on days 1 and 2 postinoculation: apoptosis shown by caspase-3 cleavage was present on days 1 and 2, while necroptosis shown by increased levels of phospho- mixed lineage kinase domain-like protein (MLKL) and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) was present on day 1 Cell death in alveolar type I (AT1) cells assessed by bronchoalveolar lavage fluid receptor for advanced glycation end points (RAGE) levels was high, yet AT2 cell death was limited while both mitochondrial biogenesis and mitophagy were induced. These mitochondrial QC mechanisms were evaluated mainly in AT2 cells by localizing increases in citrate synthase content, increases in nuclear mitochondrial biogenesis regulators nuclear respiratory factor-1 (NRF-1) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and increases in light chain 3B protein (LC3-I)/LC3II ratios. Concomitant changes in p62, Pink 1, and Parkin protein levels indicated activation of mitophagy. By confocal microscopy, mitochondrial biogenesis and mitophagy were often observed on day 1 within the same AT2 cells. These findings imply that mitochondrial QC activation in pneumonia-damaged AT2 cells promotes cell survival in support of alveolar function.
Collapse
Affiliation(s)
- Hagir B Suliman
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Bryan Kraft
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Raquel Bartz
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Lingye Chen
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Karen E Welty-Wolf
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Claude A Piantadosi
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
36
|
Radcliff FJ, Clow F, Mahadevan M, Johnston J, Proft T, Douglas RG, Fraser JD. A potential role for staphylococcal and streptococcal superantigens in driving skewing of TCR Vβ subsets in tonsillar hyperplasia. Med Microbiol Immunol 2017; 206:337-346. [DOI: 10.1007/s00430-017-0510-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/02/2017] [Indexed: 11/29/2022]
|
37
|
Parker D. Humanized Mouse Models of Staphylococcus aureus Infection. Front Immunol 2017; 8:512. [PMID: 28523002 PMCID: PMC5415562 DOI: 10.3389/fimmu.2017.00512] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/18/2017] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus is a successful human pathogen that has adapted itself in response to selection pressure by the human immune system. A commensal of the human skin and nose, it is a leading cause of several conditions: skin and soft tissue infection, pneumonia, septicemia, peritonitis, bacteremia, and endocarditis. Mice have been used extensively in all these conditions to identify virulence factors and host components important for pathogenesis. Although significant effort has gone toward development of an anti-staphylococcal vaccine, antibodies have proven ineffective in preventing infection in humans after successful studies in mice. These results have raised questions as to the utility of mice to predict patient outcome and suggest that humanized mice might prove useful in modeling infection. The development of humanized mouse models of S. aureus infection will allow us to assess the contribution of several human-specific virulence factors, in addition to exploring components of the human immune system in protection against S. aureus infection. Their use is discussed in light of several recently reported studies.
Collapse
Affiliation(s)
- Dane Parker
- Department of Pediatrics, Columbia University, New York, NY, USA
| |
Collapse
|
38
|
Bröker BM, Mrochen D, Péton V. The T Cell Response to Staphylococcus aureus. Pathogens 2016; 5:pathogens5010031. [PMID: 26999219 PMCID: PMC4810152 DOI: 10.3390/pathogens5010031] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a dangerous pathogen and a leading cause of both nosocomial and community acquired bacterial infection worldwide. However, on the other hand, we are all exposed to this bacterium, often within the first hours of life, and usually manage to establish equilibrium and coexist with it. What does the adaptive immune system contribute toward lifelong control of S. aureus? Will it become possible to raise or enhance protective immune memory by vaccination? While in the past the S. aureus-specific antibody response has dominated this discussion, the research community is now coming to appreciate the role that the cellular arm of adaptive immunity, the T cells, plays. There are numerous T cell subsets, each with differing functions, which together have the ability to orchestrate the immune response to S. aureus and hence to tip the balance between protection and pathology. This review summarizes the state of the art in this dynamic field of research.
Collapse
Affiliation(s)
- Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Daniel Mrochen
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Vincent Péton
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| |
Collapse
|
39
|
McGuinness WA, Kobayashi SD, DeLeo FR. Evasion of Neutrophil Killing by Staphylococcus aureus. Pathogens 2016; 5:E32. [PMID: 26999220 PMCID: PMC4810153 DOI: 10.3390/pathogens5010032] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 01/08/2023] Open
Abstract
Staphylococcus aureus causes many types of infections, ranging from self-resolving skin infections to severe or fatal pneumonia. Human innate immune cells, called polymorphonuclear leukocytes (PMNs or neutrophils), are essential for defense against S. aureus infections. Neutrophils are the most prominent cell type of the innate immune system and are capable of producing non-specific antimicrobial molecules that are effective at eliminating bacteria. Although significant progress has been made over the past few decades, our knowledge of S. aureus-host innate immune system interactions is incomplete. Most notably, S. aureus has the capacity to produce numerous molecules that are directed to protect the bacterium from neutrophils. Here we review in brief the role played by neutrophils in defense against S. aureus infection, and correspondingly, highlight selected S. aureus molecules that target key neutrophil functions.
Collapse
Affiliation(s)
- Will A McGuinness
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA.
| | - Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA.
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA.
| |
Collapse
|
40
|
Healthcare- and Community-Associated Methicillin-Resistant Staphylococcus aureus (MRSA) and Fatal Pneumonia with Pediatric Deaths in Krasnoyarsk, Siberian Russia: Unique MRSA's Multiple Virulence Factors, Genome, and Stepwise Evolution. PLoS One 2015; 10:e0128017. [PMID: 26047024 PMCID: PMC4457420 DOI: 10.1371/journal.pone.0128017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 04/21/2015] [Indexed: 12/25/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a common multidrug-resistant (MDR) pathogen. We herein discussed MRSA and its infections in Krasnoyarsk, Siberian Russia between 2007 and 2011. The incidence of MRSA in 3,662 subjects was 22.0% and 2.9% for healthcare- and community-associated MRSA (HA- and CA-MRSA), respectively. The 15-day mortality rates for MRSA hospital- and community-acquired pneumonia (HAP and CAP) were 6.5% and 50%, respectively. MRSA CAP cases included pediatric deaths; of the MRSA pneumonia episodes available, ≥27.3% were associated with bacteremia. Most cases of HA-MRSA examined exhibited ST239/spa3(t037)/SCCmecIII.1.1.2 (designated as ST239Kras), while all CA-MRSA cases examined were ST8/spa1(t008)/SCCmecIV.3.1.1(IVc) (designated as ST8Kras). ST239Kras and ST8Kras strongly expressed cytolytic peptide (phenol-soluble modulin α, PSMα; and δ-hemolysin, Hld) genes, similar to CA-MRSA. ST239Kras pneumonia may have been attributed to a unique set of multiple virulence factors (MVFs): toxic shock syndrome toxin-1 (TSST-1), elevated PSMα/Hld expression, α-hemolysin, the staphylococcal enterotoxin SEK/SEQ, the immune evasion factor SCIN/SAK, and collagen adhesin. Regarding ST8Kras, SEA was included in MVFs, some of which were common to ST239Kras. The ST239Kras (strain OC3) genome contained: a completely unique phage, φSa7-like (W), with no att repetition; S. aureus pathogenicity island SaPI2R, the first TSST-1 gene-positive (tst+) SaPI in the ST239 lineage; and a super copy of IS256 (≥22 copies/genome). ST239Kras carried the Brazilian SCCmecIII.1.1.2 and United Kingdom-type tst. ST239Kras and ST8Kras were MDR, with the same levofloxacin resistance mutations; small, but transmissible chloramphenicol resistance plasmids spread widely enough to not be ignored. These results suggest that novel MDR and MVF+ HA- and CA-MRSA (ST239Kras and ST8Kras) emerged in Siberian Russia (Krasnoyarsk) associated with fatal pneumonia, and also with ST239Kras, a new (Siberian Russian) clade of the ST239 lineage, which was created through stepwise evolution during its potential transmission route of Brazil-Europe-Russia/Krasnoyarsk, thereby selective advantages from unique MVFs and the MDR.
Collapse
|
41
|
Superantigens Modulate Bacterial Density during Staphylococcus aureus Nasal Colonization. Toxins (Basel) 2015; 7:1821-36. [PMID: 26008236 PMCID: PMC4448176 DOI: 10.3390/toxins7051821] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/15/2015] [Indexed: 02/06/2023] Open
Abstract
Superantigens (SAgs) are potent microbial toxins that function to activate large numbers of T cells in a T cell receptor (TCR) Vβ-specific manner, resulting in excessive immune system activation. Staphylococcus aureus possesses a large repertoire of distinct SAgs, and in the context of host-pathogen interactions, staphylococcal SAg research has focused primarily on the role of these toxins in severe and invasive diseases. However, the contribution of SAgs to colonization by S. aureus remains unclear. We developed a two-week nasal colonization model using SAg-sensitive transgenic mice expressing HLA-DR4, and evaluated the role of SAgs using two well-studied stains of S. aureus. S. aureus Newman produces relatively low levels of staphylococcal enterotoxin A (SEA), and although we did not detect significant TCR-Vβ specific changes during wild-type S. aureus Newman colonization, S. aureus Newman Δsea established transiently higher bacterial loads in the nose. S. aureus COL produces relatively high levels of staphylococcal enterotoxin B (SEB), and colonization with wild-type S. aureus COL resulted in clear Vβ8-specific T cell skewing responses. S. aureus COL Δseb established consistently higher bacterial loads in the nose. These data suggest that staphylococcal SAgs may be involved in regulating bacterial densities during nasal colonization.
Collapse
|
42
|
McLean R, Rash NL, Robinson C, Waller AS, Paillot R. Localised mitogenic activity in horses following infection with Streptococcus equi. Res Vet Sci 2015; 100:100-4. [PMID: 25841794 DOI: 10.1016/j.rvsc.2015.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 02/09/2015] [Accepted: 03/06/2015] [Indexed: 11/17/2022]
Abstract
Streptococcus equi subspecies equi (S. equi) is the causative agent of strangles, a highly contagious upper respiratory disease of equids. Streptococcus equi produces superantigens (sAgs), which are thought to contribute to strangles pathogenicity through non-specific T-cell activation and pro-inflammatory response. Streptococcus equi infection induces abscesses in the lymph nodes of the head and neck. In some individuals, some abscess material remains into the guttural pouch and inspissates over time to form chondroids which can harbour live S. equi. The aim of this study was to determine the sites of sAg production during infection and therefore improve our understanding of their role. Abscess material, chondroids and serum collected from Equidae with signs of strangles were tested in mitogenic assays. Mitogenic sAg activity was only detected in abscess material and chondroids. Our data support the localised in vivo activity of sAg during both acute and carrier phases of S. equi infection.
Collapse
Affiliation(s)
- R McLean
- Animal Health Trust, Centre of Preventative Medicine, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK
| | - N L Rash
- Animal Health Trust, Centre of Preventative Medicine, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK
| | - C Robinson
- Animal Health Trust, Centre of Preventative Medicine, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK
| | - A S Waller
- Animal Health Trust, Centre of Preventative Medicine, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK
| | - R Paillot
- Animal Health Trust, Centre of Preventative Medicine, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK.
| |
Collapse
|
43
|
Cuny C, Abdelbary M, Layer F, Werner G, Witte W. Prevalence of the immune evasion gene cluster in Staphylococcus aureus CC398. Vet Microbiol 2015; 177:219-23. [PMID: 25778546 DOI: 10.1016/j.vetmic.2015.02.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/21/2015] [Accepted: 02/23/2015] [Indexed: 10/23/2022]
Abstract
The immune evasion gene cluster (IEC) is typical for Staphylococcus aureus isolated from humans but is usually absent in S. aureus isolated from animals. Previous studies have shown that methicillin resistant S. aureus (MRSA) CC398 obviously lost the IEC when evolving as livestock-associated MRSA from a human-adapted, methicillin-susceptible ancestor. This study aimed to look for the presence of IEC in MRSA from pigs and horses as well as from the colonization of humans with occupational animal contact and from infections in humans. For comparison, methicillin susceptible S. aureus (MSSA) isolates from infections in humans were included. We did not detect the IEC among 94 isolates from the nasal colonization of pigs; however, the IEC was found in 6 of 61 isolates from nosocomial infections in horses. MRSA CC398 isolates from the nasal colonization of 138 pig farmers were negative for the IEC. It was detected, however, in 4 of 69 veterinarians treating horses. Among 99 epidemiologically unrelated MRSA isolates attributed to CC398 originating from infections in humans, 19 were positive for the IEC. Only three of these isolates which also contained luk-PV were attributed to the ancestral, human-adapted subpopulation of CC398 by means of PCR for detection of canonical SNPs. A considerable proportion of LA-MRSA CC398 attributed to the animal subpopulation and originating from infections in humans had acquired the IEC; this acquisition is, however, obviously not a prerequisite to the capacity of LA-MRSA CC398 to cause infections in this host. Among 15 MSSA CC398 isolates from infections in humans, 11 contained the IEC, and of these, two were attributed to the animal subpopulation. Six isolates containing both the IEC and luk-PV were attributed to the ancestral, human subpopulation. Re-acquisition of the IEC by LA-MRSA CC398 suggests readaptation to the human host. In epidemiological surveillance, discrimination from the ancestral human subpopulation is important.
Collapse
Affiliation(s)
- Christiane Cuny
- Robert Koch Institute, Wernigerode Branch, Burgstrasse 37, 38855 Wernigerode, Germany
| | - Mohamed Abdelbary
- Robert Koch Institute, Wernigerode Branch, Burgstrasse 37, 38855 Wernigerode, Germany
| | - Franziska Layer
- Robert Koch Institute, Wernigerode Branch, Burgstrasse 37, 38855 Wernigerode, Germany
| | - Guido Werner
- Robert Koch Institute, Wernigerode Branch, Burgstrasse 37, 38855 Wernigerode, Germany
| | - Wolfgang Witte
- Robert Koch Institute, Wernigerode Branch, Burgstrasse 37, 38855 Wernigerode, Germany.
| |
Collapse
|
44
|
Guimarães MA, Ramundo MS, Américo MA, de Mattos MC, Souza RR, Ramos-Júnior ES, Coelho LR, Morrot A, Melo PA, Fracalanzza SEL, Ferreira FA, Figueiredo AMS. A comparison of virulence patterns and in vivo fitness between hospital- and community-acquired methicillin-resistant Staphylococcus aureus related to the USA400 clone. Eur J Clin Microbiol Infect Dis 2015; 34:497-509. [PMID: 25311987 DOI: 10.1007/s10096-014-2253-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 09/16/2014] [Indexed: 10/24/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) isolates genetically related to the CA-MRSA clone MW2/USA400 (ST1-SCCmecIV lineage) from the United States have emerged in hospitals in Rio de Janeiro and are associated with nosocomial bloodstream infections. To understand the virulence mechanisms involved in the adaptability of ST1 isolates as a hospital pathogen in Rio de Janeiro, we compared the virulence traits and fitness properties of the Brazilian isolates with those displayed by the CA-MRSA isolates from the United States. Similar to the USA400 from the United States, all the Brazilian isolates tested carried the genes encoding SEH and LukDE. In contrast, none of the Brazilian isolates carried the lukSF PVL, sea, sec, and sek genes. Competition experiments in mice demonstrated a significant increase in the fitness for the CA-MRSA isolates MW2 and USA400-0051 from the United States compared to other isolates. In the foreign body animal model, 83 % more North-American bacterial cells were recovered compared to the Brazilian ST1 isolates. Differences in gene expression of important virulence factors were detected. Transcription of rnaIII and psmα3 was increased about two-fold in the isolates from the United States, and sasG about two-fold in the Brazilian isolates. Thus, it is possible that the virulence attenuation observed among the Brazilian hospital isolates, associated with the acquisition of multiple resistant determinants, are consequences of microevolutionary events that contributed to the necessary fitness adjustment of this lineage, allowing a typically community-acquired MRSA (MW2/USA400) to emerge as a successful hospital pathogen (Brazilian ST1-SCCmecIV).
Collapse
Affiliation(s)
- M A Guimarães
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Koymans KJ, Vrieling M, Gorham RD, van Strijp JAG. Staphylococcal Immune Evasion Proteins: Structure, Function, and Host Adaptation. Curr Top Microbiol Immunol 2015; 409:441-489. [PMID: 26919864 DOI: 10.1007/82_2015_5017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Staphylococcus aureus is a successful human and animal pathogen. Its pathogenicity is linked to its ability to secrete a large amount of virulence factors. These secreted proteins interfere with many critical components of the immune system, both innate and adaptive, and hamper proper immune functioning. In recent years, numerous studies have been conducted in order to understand the molecular mechanism underlying the interaction of evasion molecules with the host immune system. Structural studies have fundamentally contributed to our understanding of the mechanisms of action of the individual factors. Furthermore, such studies revealed one of the most striking characteristics of the secreted immune evasion molecules: their conserved structure. Despite high-sequence variability, most immune evasion molecules belong to a small number of structural categories. Another remarkable characteristic is that S. aureus carries most of these virulence factors on mobile genetic elements (MGE) or ex-MGE in its accessory genome. Coevolution of pathogen and host has resulted in immune evasion molecules with a highly host-specific function and prevalence. In this review, we explore how these shared structures and genomic locations relate to function and host specificity. This is discussed in the context of therapeutic options for these immune evasion molecules in infectious as well as in inflammatory diseases.
Collapse
Affiliation(s)
- Kirsten J Koymans
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Manouk Vrieling
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ronald D Gorham
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
46
|
Staphylococcus aureus secretes a unique class of neutrophil serine protease inhibitors. Proc Natl Acad Sci U S A 2014; 111:13187-92. [PMID: 25161283 DOI: 10.1073/pnas.1407616111] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Neutrophils are indispensable for clearing infections with the prominent human pathogen Staphylococcus aureus. Here, we report that S. aureus secretes a family of proteins that potently inhibits the activity of neutrophil serine proteases (NSPs): neutrophil elastase (NE), proteinase 3, and cathepsin G. The NSPs, but not related serine proteases, are specifically blocked by the extracellular adherence protein (Eap) and the functionally orphan Eap homologs EapH1 and EapH2, with inhibitory-constant values in the low-nanomolar range. Eap proteins are together essential for NSP inhibition by S. aureus in vitro and promote staphylococcal infection in vivo. The crystal structure of the EapH1/NE complex showed that Eap molecules constitute a unique class of noncovalent protease inhibitors that occlude the catalytic cleft of NSPs. These findings increase our insights into the complex pathogenesis of S. aureus infections and create opportunities to design novel treatment strategies for inflammatory conditions related to excessive NSP activity.
Collapse
|