201
|
Transcriptional regulation of virulence factors Hla and phenol-soluble modulins α by AraC-type regulator Rbf in Staphylococcus aureus. Int J Med Microbiol 2020; 310:151436. [PMID: 32654771 DOI: 10.1016/j.ijmm.2020.151436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/25/2020] [Accepted: 06/14/2020] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is a gram-positive pathogenic bacterium and is capable of secreting numerous toxins interfering directly with the host to cause acute infections. Rbf, a transcriptional regulator of AraC/XylS family, has been reported to promote biofilm formation in polysaccharide intercellular adhesion (PIA) mediated manner to cause chronic infections. In this study, we revealed the new virulence-mediated role of Rbf that can negatively regulate the hemolytic activity. Furthermore, Rbf can specifically bind to the hla and psmα promoters to repress their expression, resulting in significantly decreased production of phenol-soluble modulins α (PSMα) and alpha-toxin. Accordingly, the rbf mutant strain exhibited the increased pathogenicity compared to the wild-type (WT) strain in a mouse subcutaneous abscess model, representing a type of acute infection by S. aureus. Collectively, our results provide a novel insight into the virulence regulation and acute infections mediated by Rbf in S. aureus.
Collapse
|
202
|
Vijayakumar K, Bharathidasan V, Manigandan V, Jeyapragash D. Quebrachitol inhibits biofilm formation and virulence production against methicillin-resistant Staphylococcus aureus. Microb Pathog 2020; 149:104286. [PMID: 32502632 DOI: 10.1016/j.micpath.2020.104286] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/12/2020] [Accepted: 05/26/2020] [Indexed: 01/30/2023]
Abstract
The present study evaluated the quebrachitol (QBC) antibiofilm and antivirulence potential against methicillin-resistant Staphylococcus aureus (MRSA). QBC inhibited MRSA biofilm formation at concentration dependent manner without affecting the bacterial growth. Then, QBC biofilm efficacy was confirmed with light and confocal laser scanning microscopy analysis. QBC treatment significantly inhibited the biofilm formation on stainless steel, titanium and silicone surfaces. Besides, QBC treatment significantly reduced the MRSA virulence productions such as lipase and hemolysis. Moreover, it reduced MRSA survival rate in the presence of hydrogen peroxide. QBC treatment inhibited the MRSA adherence on hydrophobic, hydrophilic, collagen coating and fibrinogen coating surfaces. As well as it significantly reduced the autolysin and bacterial aggregation progress. The real-time PCR analysis revealed the ability of QBC downregulated the virulence genes expression including global regulator sarA, agr and polysaccharide intracellular adhesion (PIA) encode ica. The cumulative results of the present study suggest that QBC as a potential agent to combat against MRSA pathogenesis.
Collapse
Affiliation(s)
- Karuppiah Vijayakumar
- Center of Advanced Study in Marine Biology, Annamalai University, Parangipettai, 608 502, Tamil Nadu, India.
| | - Veeraiyan Bharathidasan
- Center of Advanced Study in Marine Biology, Annamalai University, Parangipettai, 608 502, Tamil Nadu, India
| | - Vajravelu Manigandan
- Center of Advanced Study in Marine Biology, Annamalai University, Parangipettai, 608 502, Tamil Nadu, India
| | - Danaraj Jeyapragash
- Center of Advanced Study in Marine Biology, Annamalai University, Parangipettai, 608 502, Tamil Nadu, India
| |
Collapse
|
203
|
Song Y, Xu M, Li Y, Li Y, Gu W, Halimu G, Fu X, Zhang H, Zhang C. An iRGD peptide fused superantigen mutant induced tumor-targeting and T lymphocyte infiltrating in cancer immunotherapy. Int J Pharm 2020; 586:119498. [PMID: 32505575 DOI: 10.1016/j.ijpharm.2020.119498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/07/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022]
Abstract
Solid tumors are intrinsically resistant to immunotherapy because of the major challenges including the immunosuppression and poor penetration of drugs and lymphocytes into solid tumors due to the complicated tumor microenvironment (TME). Our previous study has created a novel superantigen mutant ST-4 to efficiently active the T lymphocytes and alleviate immune suppression. In the present study, to accumulate ST-4 into the TME, we constructed a recombinant protein, ST-4-iRGD, by fusing ST-4 to a tumor-homing peptide, iRGD. We hypothesized that ST-4-iRGD could internalize into the TME through iRGD-mediated tumor targeting and tumor tissue penetrating to activate the regional immunoreaction. The results of in vitro studies showed that ST-4-iRGD achieved improved tumor targeting and cytotoxicity in mouse B16F10 melanoma cells. The iRGD-mediated tumor tissue penetration was further confirmed by imaging and immunofluorescence studies in vivo, wherein higher distribution of ST-4-iRGD was observed in the mouse 4T1 breast tumor model. Moreover, ST-4-iRGD exhibited enhanced anti-solid tumor characteristics and induced improved lymphocyte infiltration in the B16F10 and 4T1 models. In conclusion, using iRGD to facilitate better dissemination of the therapeutic agent ST-4 throughout a solid tumor mass is feasible, and ST-4-iRGD may be a potential candidate for efficient cancer immunotherapy in the future.
Collapse
Affiliation(s)
- Yubo Song
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Mingkai Xu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China.
| | - Yongqiang Li
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Yansheng Li
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Wu Gu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Gulinare Halimu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Xuanhe Fu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Huiwen Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Chenggang Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| |
Collapse
|
204
|
Houri H, Samadpanah M, Tayebi Z, Norouzzadeh R, Malekabad ES, Dadashi AR. Investigating the toxin profiles and clinically relevant antibiotic resistance genes among Staphylococcus aureus isolates using multiplex-PCR assay in Tehran, Iran. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
205
|
Goudarzi M, Razeghi M, Hashemi A, Pouriran R, Dadashi M, Tayebi Z. Genetic analysis of toxic shock syndrome toxin (TST) positive Staphylococcus aureus strains isolated from wound infections in Tehran hospitals, Iran. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
206
|
Abstract
In the 1980s, menstrual toxic shock syndrome (mTSS) became a household topic, particularly among mothers and their daughters. The research performed at the time, and for the first time, exposed the American public as well as the biomedical community, in a major way, to understanding disease progression and investigation. Those studies led to the identification of the cause, Staphylococcus aureus and the pyrogenic toxin superantigen TSS toxin 1 (TSST-1), and many of the risk factors, for example, tampon use. Those studies in turn led to TSS warning labels on the outside and inside of tampon boxes and, as important, uniform standards worldwide of tampon absorbency labeling. This review addresses our understanding of the development and conclusions related to mTSS and risk factors. We leave the final message that even though mTSS is not commonly in the news today, cases continue to occur. Additionally, S. aureus strains cycle in human populations in roughly 10-year intervals, possibly dependent on immune status. TSST-1-producing S. aureus bacteria appear to be reemerging, suggesting that physician awareness of this emergence and mTSS history should be heightened.
Collapse
|
207
|
Shang Y, Wang X, Chen Z, Lyu Z, Lin Z, Zheng J, Wu Y, Deng Q, Yu Z, Zhang Y, Qu D. Staphylococcus aureus PhoU Homologs Regulate Persister Formation and Virulence. Front Microbiol 2020; 11:865. [PMID: 32670206 PMCID: PMC7326077 DOI: 10.3389/fmicb.2020.00865] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
PhoU homologs are one of the determinant factors in the regulation of persister formation and phosphate metabolism in many bacterial species; however, the functions of PhoU homologs exhibit species-specific characteristics. The pathogenesis of Staphylococcus aureus is closely correlated with persister formation and virulence factors. The functions of two PhoU homologs, PhoU1 and PhoU2, in S. aureus are unclear yet. In this study, single- and double-deletion mutants of phoU1 and phoU2 were generated in strain USA500 2395. The ΔphoU1 or ΔphoU2 mutants displayed a change in persister formation and virulence compared to the parent strain; the persisters to vancomycin and levofloxacin were decreased at least 1,000-fold, and the number of intracellular bacteria surviving in the A549 cells for 24 h decreased to 82 or 85%. The α-hemolysin expression and activity were increased in the ΔphoU2 mutants. Transcriptome analysis revealed that 573 or 285 genes were differentially expressed by at least 2.0-fold in the ΔphoU1 or ΔphoU2 mutant vs. the wild type. Genes involved in carbon and pyruvate metabolism were up-regulated, and virulence genes and virulence regulatory genes were down-regulated, including type VII secretion system, serine protease, leukocidin, global regulator (sarA, rot), and the two-component signal transduction system (saeS). Correspondingly, the deletion of the phoU1 or phoU2 resulted in increased levels of intracellular pyruvate and ATP. Deletion of the phoU2, but not the phoU1, resulted in the up-regulation of inorganic phosphate transport genes and increased levels of intracellular inorganic polyphosphate. In conclusion, both PhoU1 and PhoU2 in S. aureus regulate virulence by the down-regulation of multiple virulence factors (type VII secretion system, serine protease, and leucocidin) and the persister generation by hyperactive carbon metabolism accompanied by increasing intracellular ATP. The results in S. aureus are different from what we have previously found in Staphylococcus epidermis, where only PhoU2 regulates biofilm and persister formation. The different functions of PhoU homologs between the two species of Staphylococcus warrant further investigation.
Collapse
Affiliation(s)
- Yongpeng Shang
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaofei Wang
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhong Chen
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhihui Lyu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhiwei Lin
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Jinxin Zheng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiwen Deng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Zhijian Yu
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
208
|
Virulence Characteristics of mecA-Positive Multidrug-Resistant Clinical Coagulase-Negative Staphylococci. Microorganisms 2020; 8:microorganisms8050659. [PMID: 32369929 PMCID: PMC7284987 DOI: 10.3390/microorganisms8050659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
Coagulase-negative staphylococci (CoNS) are an important group of opportunistic pathogenic microorganisms that cause infections in hospital settings and are generally resistant to many antimicrobial agents. We report on phenotypic and genotypic virulence characteristics of a select group of clinical, mecA-positive (encoding penicillin-binding protein 2a) CoNS isolates. All CoNS were resistant to two or more antimicrobials with S. epidermidis strain 214EP, showing resistance to fifteen of the sixteen antimicrobial agents tested. Aminoglycoside-resistance genes were the ones most commonly detected. The presence of megaplasmids containing both horizontal gene transfer and antimicrobial resistance genetic determinants indicates that CoNS may disseminate antibiotic resistance to other bacteria. Staphylococcus sciuri species produced six virulence enzymes, including a DNase, gelatinase, lipase, phosphatase, and protease that are suspected to degrade tissues into nutrients for bacterial growth and contribute to the pathogenicity of CoNS. The PCR assay for the detection of biofilm-associated genes found the eno (encoding laminin-binding protein) gene in all isolates. Measurement of their biofilm-forming ability and Spearman’s rank correlation coefficient analyses revealed that the results of crystal violet (CV) and extracellular polymeric substances (EPS) assays were significantly correlated (ρ = 0.9153, P = 3.612e-12). The presence of virulence factors, biofilm-formation capability, extracellular enzymes, multidrug resistance, and gene transfer markers in mecA-positive CoNS clinical strains used in this study makes them powerful opportunistic pathogens. The study also warrants a careful evaluation of nosocomial infections caused by CoNS and may be useful in studying the mechanism of virulence and factors associated with their pathogenicity in vivo and developing effective strategies for mitigation.
Collapse
|
209
|
Zhou P, Chen J, Li HH, Sun J, Gao SX, Zheng QW, Wei L, Jiang CY, Guan JC. Exposure of pregnant rats to staphylococcal enterotoxin B attenuates the response of increased Tregs to re-exposure to SEB in the thymus of adult offspring. Microb Pathog 2020; 145:104225. [PMID: 32353581 DOI: 10.1016/j.micpath.2020.104225] [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: 02/28/2020] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 11/17/2022]
Abstract
Regulatory T cells (Tregs) play an essential role during homeostasis and tolerance of the immune system. Based on our previous study that exposure of pregnant rats to staphylococcal enterotoxin B (SEB) can alter the percentage of CD4/CD8 subsets in the thymus of the offspring, in this study, we focus on the influence of exposure of pregnant rats to SEB on number, function and response of Tregs in the thymus of the offspring. Pregnant rats at gestational day of 16 were intravenously injected with 15 μg SEB and the thymuses of the neonatal and adult offspring were harvested for this study. We found that exposure of pregnant rats to SEB could significantly increase the absolute number of Tregs and the FoxP3 expression level in the thymus of not only neonatal but also adult offspring. Re-exposure of adult offspring to SEB remarkably reduced the suppressive capacity of Tregs to CD4+ T cells and the expression levels of TGF-β and IL-10 in the thymus, but had no effect on production of IL-4 and IFN-γ. Furthermore, it also notedly decreased the absolute number of Tregs and the FoxP3 expression level. These data suggest that prenatal exposure of pregnant rats to SEB attenuates the response of increased Tregs to re-exposure to SEB in the thymus of adult offspring.
Collapse
Affiliation(s)
- Ping Zhou
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jie Chen
- Department of Cardiology, Jiande Branch, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiande, 311600, PR China
| | - Hui-Hui Li
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jing Sun
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Shu-Xian Gao
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Qing-Wei Zheng
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Li Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Cheng-Yi Jiang
- Department of Otolaryngology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233033, PR China
| | - Jun-Chang Guan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| |
Collapse
|
210
|
Fan E, Peng J, Shi Y, Ouyang H, Xu Z, Fu Z. Quantification of live Gram-positive bacteria via employing artificial antibacterial peptide-coated magnetic spheres as isolation carriers. Microchem J 2020. [DOI: 10.1016/j.microc.2020.104643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
211
|
Khateb H, Klös G, Meyer RL, Sutherland DS. Development of a Label-Free LSPR-Apta Sensor for Staphylococcus aureus Detection. ACS APPLIED BIO MATERIALS 2020; 3:3066-3077. [DOI: 10.1021/acsabm.0c00110] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Heba Khateb
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Gunnar Klös
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Rikke L. Meyer
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Duncan S. Sutherland
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| |
Collapse
|
212
|
Effect of (-)-Epigallocatechin Gallate to Staphylococcal Enterotoxin A on Toxin Activity. Molecules 2020; 25:molecules25081867. [PMID: 32316678 PMCID: PMC7221706 DOI: 10.3390/molecules25081867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 01/16/2023] Open
Abstract
Staphylococcal enterotoxin A (SEA) functions both as superantigens that stimulate non-specific T cell proliferation as well as potent gastrointestinal toxins. We previously reported that (-)-epigallocatechin gallate (EGCG) binds to SEA. Therefore, the ability of EGCG to inhibit SEA toxin activity was examined. As a result, EGCG significantly decreased SEA-induced expression and production of interferon gamma (IFN-γ). In addition, EGCG inhibited SEA-induced spleen cell proliferation. To investigate the role of the galloyl group in EGCG on SEA cytotoxicity in more detail, the effect of the binding of a hydroxyl group at position 3 of the galloyl group in EGCG to SEA on SEA cytotoxicity was examined using two methylated EGCG. SEA cytotoxicity was significantly controlled in both (-)-3''-Me-EGCG and (-)-4''-Me-EGCG. These results suggest that EGCG inhibits toxic activity via direct interaction with SEA or without any interaction with SEA. The binding affinity between SEA and EGCG under in vivo conditions was examined using a model solution. Although after treatment under acidic and alkaline conditions, the presence of protein and the digestive tract model solution, EGCG still interacted with SEA. Our studies are the first to demonstrate the effect of the binding of EGCG to SEA on toxin activity.
Collapse
|
213
|
Liu Y, Song Z, Ge S, Zhang J, Xu L, Yang F, Lu D, Luo P, Gu J, Zou Q, Zeng H. Determining the immunological characteristics of a novel human monoclonal antibody developed against staphylococcal enterotoxin B. Hum Vaccin Immunother 2020; 16:1708-1718. [PMID: 32275466 DOI: 10.1080/21645515.2020.1744362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Staphylococci are the main cause of nosocomial infections globally. The exotoxin staphylococcal enterotoxin B (SEB) produced by methicillin-resistant Staphylococcus aureus is a major cause of pathology after a staphylococcal infection. We previously isolated an anti-SEB human monoclonal antibody designated as M0313. Here we further characterize this antibody in vitro and in vivo. Immunoblotting analysis and ELISA results indicated that M0313 accurately recognized and bound to SEB. Its binding affinity to native SEB was measured at the low nM level. M0313 effectively inhibited SEB from inducing mouse splenic lymphocyte and human peripheral blood mononuclear cell proliferation and cytokine release in cell culture. M0313 also neutralized SEB toxicity in BALB/c female mice. Most importantly, M0313 promoted the survival of mice treated with SEB-expressing bacteria. In-vivo imaging revealed that M0313 treatment significantly reduced the replication of SEB-expressing bacteria in mice. The neutralization capacity of M0313 correlated with its ability to block SEB from binding to major histocompatibility complex II and T-cell receptor by binding to the SEB residues 85-102 and 90-92. Thus, the monoclonal antibody M0313 may be developed into a therapeutic agent.
Collapse
Affiliation(s)
- Yuanyuan Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Zhen Song
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China.,Clinical Laboratory Department, Army 954th Hospital, General Hospital of Tibet Military Region , Tibet, PR China
| | - Shuang Ge
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Limin Xu
- Research and Development Department, Chengdu Olymvax Biotechnology Co., Ltd ., Chengdu, Sichuan, PR China
| | - Feng Yang
- Research and Development Department, Chengdu Olymvax Biotechnology Co., Ltd ., Chengdu, Sichuan, PR China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Jiang Gu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| |
Collapse
|
214
|
Vaughn JM, Abdi RD, Gillespie BE, Kerro Dego O. Genetic diversity and virulence characteristics of Staphylococcus aureus isolates from cases of bovine mastitis. Microb Pathog 2020; 144:104171. [PMID: 32224210 DOI: 10.1016/j.micpath.2020.104171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Staphylococcus aureus is one of the major bacterial mastitis pathogens with significant effects on animal and human health. Some studies showed that S. aureus strains that infect different host species are genetically distinct, although most strains can infect a wide range of host species. However, there are no clearly defined clonal patterns of S. aureus strains that are known to infect a specific host. The objectives of this study were to evaluate the clonal diversity and virulence characteristics of S. aureus isolates from cases of bovine mastitis. Bacteriological tests were conducted on milk samples from cases of bovine mastitis from 11 dairy farms including some milk samples from unknown farms in Eastern Tennessee. Overall, a total of 111 S. aureus were isolated and identified, and further evaluated for their genetic diversity by pulsed-field gel electrophoresis (PFGE) and virulence characteristics by PCR. Genotypic virulence factors including staphylococcal enterotoxins, and toxic shock syndrome toxin 1 (tsst-1) were tested by PCR. In addition, the association among several known virulence factors of these isolates based on our current and previous studies in our lab were evaluated. Previously generated data that were included in the analysis of association among virulence factors were the presence of biofilm production associated genes in the ica operon such as icaA, icaD and icaAB, and phenotypic virulence characteristics such as hemolysis on blood agar, slime production and resistance or susceptibility to ten commonly used antimicrobials in dairy farms. The PFGE results showed the presence of 16 PFGE types (designated A - P) throughout farms, of which three pulsotypes, I, M and O were the most frequently isolated PFGE types from most farms. The PFGE type M was the most prevalent of all 16 PFGE types, with 64 isolates being present among nine farms. The PCR results of enterotoxin genes showed that out of the total 111 tested 84 (75.7%) were negative whereas 13 (11.7%), 2 (1.8%), 3 (2.7%), 1 (0.9%) and 8 (7.2%) were positive for seb, seb and sec, sec, see, and tsst-1, respectively. All 111 isolates were negative for sea and sej. Results of the evaluation of I, M and O strains adhesion to and invasion into mammary epithelial cells showed that the total count of each strain of bacteria adhered to and invaded into mammary epithelial cell line (MAC-T cells) was not significantly different (P > 0.05). This may be an indication that there is no significant difference in their ability to establish early host-pathogen interaction and colonization of the host. There were no statistically significant associations among PFGE types and other known virulence factors of these strains. However, PFGE types O and M tend to cluster with β-hemolysin, absence of enterotoxins and susceptibility to antimicrobials. In conclusion, there was not any association between pulsotype and genotypic and phenotypic virulence factors. S. aureus isolates from cases of bovine mastitis had diverse genotypes that possessed variable virulence factors.
Collapse
Affiliation(s)
- Jacqueline M Vaughn
- The University of Tennessee, Department of Animal Science, Knoxville, TN, 37996, USA
| | - Reta Duguma Abdi
- The University of Tennessee, Department of Animal Science, Knoxville, TN, 37996, USA; Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Greenvale, NY, 11548, USA
| | | | - Oudessa Kerro Dego
- The University of Tennessee, Department of Animal Science, Knoxville, TN, 37996, USA.
| |
Collapse
|
215
|
Kitadokoro K, Tanaka M, Hikima T, Okuno Y, Yamamoto M, Kamitani S. Crystal structure of pathogenic Staphylococcus aureus lipase complex with the anti-obesity drug orlistat. Sci Rep 2020; 10:5469. [PMID: 32214208 PMCID: PMC7096528 DOI: 10.1038/s41598-020-62427-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/11/2020] [Indexed: 12/26/2022] Open
Abstract
Staphylococcus aureus lipase (SAL), a triacylglycerol esterase, is an important virulence factor and may be a therapeutic target for infectious diseases. Herein, we determined the 3D structure of native SAL, the mutated S116A inactive form, and the inhibitor complex using the anti-obesity drug orlistat to aid in drug development. The determined crystal structures showed a typical α/β hydrolase motif with a dimeric form. Fatty acids bound near the active site in native SAL and inactive S116A mutant structures. We found that orlistat potently inhibits SAL activity, and it covalently bound to the catalytic Ser116 residue. This is the first report detailing orlistat–lipase binding. It provides structure-based information on the production of potent anti-SAL drugs and lipase inhibitors. These results also indicated that orlistat can be repositioned to treat bacterial diseases.
Collapse
Affiliation(s)
- Kengo Kitadokoro
- Faculty of Molecular Chemistry and Engineering, Graduate School of Science and Technology, Kyoto Institute of Technology, Hashigami-cho, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan.
| | - Mutsumi Tanaka
- Faculty of Molecular Chemistry and Engineering, Graduate School of Science and Technology, Kyoto Institute of Technology, Hashigami-cho, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Takaaki Hikima
- SR Life Science Instrumentation Team, Life Science Research Infrastructure Group, Advanced Photon Technology Division, RIKEN SPring-8 Center, 1-1-1, Koto, Sayo-cho, Sayo-gun, Hyogo, 679-6148, Japan
| | - Yukiko Okuno
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Masaki Yamamoto
- SR Life Science Instrumentation Team, Life Science Research Infrastructure Group, Advanced Photon Technology Division, RIKEN SPring-8 Center, 1-1-1, Koto, Sayo-cho, Sayo-gun, Hyogo, 679-6148, Japan
| | - Shigeki Kamitani
- Graduate School of Comprehensive Rehabilitation, College of Health and Human Sciences, Osaka Prefecture University, 3-7-30 Habikino, Habikino, 583-8555, Osaka, Japan
| |
Collapse
|
216
|
Atypical Presentation of Methicillin-Susceptible Staphylococcus aureus Infection in a Dengue-Positive Patient: A Case Report with Virulence Genes Analysis. Pathogens 2020; 9:pathogens9030190. [PMID: 32150854 PMCID: PMC7157556 DOI: 10.3390/pathogens9030190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/26/2022] Open
Abstract
Concurrent bacteraemia in patients with dengue fever is rarely reported. We report a case of a patient who initially presented with symptoms typical of dengue fever but later succumbed to septic shock caused by hypervirulent methicillin-susceptible Staphylococcus aureus (MSSA). A 50-year-old female patient with hypertension and diabetes mellitus presented with typical symptoms of dengue fever. Upon investigation, the patient reported having prolonged fever for four days prior to hospitalization. Within 24 hours post-admission, the patient developed pneumonia and refractory shock, and ultimately succumbed to multiple-organs failure. Microbiological examination of the blood culture retrieved a pan susceptible MSSA strain. Genomic sequence analyses of the MSSA strain identified genes encoding staphylococcal superantigens (enterotoxin staphylococcal enterotoxin C 3 (SEC3) and enterotoxin-like staphylococcal enterotoxins-like toxin L (SElL)) that have been associated with toxic shock syndrome in human hosts. Genes encoding important toxins (Panton-Valentine leukocidins, alpha-haemolysin, protein A) involved in the development of staphylococcal pneumonia were also present in the MSSA genome. Staphylococcus aureus co-infections in dengue are uncommon but could be exceptionally fatal if caused by a toxin-producing strain. Clinicians should be aware of the risks and signs of sepsis in dengue fever, thus allowing early diagnosis and starting of antibiotic treatment in time to lower the mortality and morbidity rates.
Collapse
|
217
|
Abstract
Peritoneal dialysis has a high acceptance rate in Latin America, thus the knowledge concerning complication patterns is of great relevance. This work reviews Latin American data on peritonitis, the most serious complication of peritoneal dialysis. The incidence of peritonitis has been reduced over time, concomitantly with the incorporation of safer exchange systems and the use of prophylactic measurements. Today, rates lower than 1 episode per 24 patient-months are commonly reported. Furthermore, changes in causative organisms have been observed, with predominance of Staphylococcus aureus up through the mid-1990s, as well as increases in coagulase-negative staphylococcus and participation of gram negatives. However, the prevalence of S. aureus is still high, due possibly to climatic conditions and the elevated prevalence of carriers. Resolution rate varies from 55% to 78%, transfer to hemodialysis from 10.9% to 15.4%, and death in 3% to 9.9% of cases. Outcome is worse in S. aureus episodes compared to those with coagulase-negative staphylococcus, despite the higher percentage of oxacillin-resistant strains among the former. In general, despite socioeconomic or climatic conditions, our results are similar to those in developed countries, perhaps as a consequence of technological improvements and/or center expertise.
Collapse
Affiliation(s)
- Pasqual Barretti
- Department of Internal Medicine,
University Hospital, Botucatu School of Medicine, UNESP, São Paulo
| | - Kleyton A. Bastos
- Department of Medicine, Federal
University of Sergipe, Sergipe, Brazil
| | - Jorge Dominguez
- Dialysis and Transplantation Service,
Miguel Perez Carreño Hospital, Caracas, Venezuela
| | - Jacqueline C.T. Caramori
- Department of Internal Medicine,
University Hospital, Botucatu School of Medicine, UNESP, São Paulo
| |
Collapse
|
218
|
Machado V, Pardo L, Cuello D, Giudice G, Luna PC, Varela G, Camou T, Schelotto F. Presence of genes encoding enterotoxins in Staphylococcus aureus isolates recovered from food, food establishment surfaces and cases of foodborne diseases. Rev Inst Med Trop Sao Paulo 2020; 62:e5. [PMID: 32049256 PMCID: PMC7014549 DOI: 10.1590/s1678-9946202062005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/06/2019] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to describe the microbiological characteristics and profile of genes encoding enterotoxins in 95 Staphylococcus aureus isolates obtained between April 2011 and December 2014 from foodstuffs, persons and surfaces of retail food stores. After microbiological identification and antimicrobial susceptibility testing, polymerase chain reactions (PCR) were performed, targeting sea, seb, sec, sed and see genes that code for classical enterotoxins (ET) A-E, and three additional genes: seg , seh and sei , coding for so-called "new enterotoxins" G, H and I. The isolates were characterized by Pulsed Field Gel Electrophoresis (PFGE), and five selected isolates were further analyzed through Multi Locus Sequence Typing (MLST). It is noteworthy that 54.7% of the examined isolates harbored one or more of the investigated ET gene types. Most positive isolates carried more than one ET gene up to five types; seg was the most frequent ET gene, followed by sei. Five enterotoxin-coding isolates also coded for some antimicrobial resistance genes. Two of them, and four additional non-enterotoxic isolates carried erm genes expressing inducible clindamycin resistance. PFGE-types were numerous and diverse, even among enterotoxin-coding strains, because most isolates did not belong to known foodborne outbreaks and the sampling period was long. MLST profiles were also varied, and a new ST 3840 was described within this species. ST 88 and ST 72 enterotoxin-coding isolates have been identified in other regions in association with foodborne outbreaks. This manuscript reports the first systematic investigation of enterotoxin genes in S. aureus isolates obtained from foodstuffs and infected people in Uruguay.
Collapse
Affiliation(s)
- Virginia Machado
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Lorena Pardo
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Dianna Cuello
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Guillermina Giudice
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Patricia Correa Luna
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Gustavo Varela
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| | - Teresa Camou
- Departamento de Laboratorios de Salud Pública, Montevideo, Uruguay
| | - Felipe Schelotto
- Universidad de la República, Facultad de Medicina, Instituto de Higiene, Departamento de Bacteriología y Virología, Montevideo, Uruguay
| |
Collapse
|
219
|
Jiang X, Yan X, Gu S, Yang Y, Zhao L, He X, Chen H, Ge J, Liu D. Biosurfactants of Lactobacillus helveticus for biodiversity inhibit the biofilm formation of Staphylococcus aureus and cell invasion. Future Microbiol 2020; 14:1133-1146. [PMID: 31512521 DOI: 10.2217/fmb-2018-0354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: This study aimed to evaluate the differences of biosurfactants produced by two Lactobacillus helveticus strains against the biofilm formation of Staphylococcus aureus in vitro and in vivo. Materials & methods: Scanning electron microscopy, Real time-quantitative PCR (RT-qPCR) and cell assay were used to analyze the inhibiting effect of biosurfactants against biofilm formation. Results & conclusion: Results showed that the biosurfactants have anti-adhesive and inhibiting effects on biofilm formation in vivo and in vitro. The biofilm-formative genes and autoinducer-2 signaling regulated these characteristics, and the biosurfactant L. helveticus 27170 is better than that of 27058. Host cell adhesion and invasion results indicated that the biosurfactants L. helveticus prevented the S. aureus invading the host cell, which may be a new strategy to eliminate biofilms.
Collapse
Affiliation(s)
- Xinpeng Jiang
- Heilongjiang Key Laboratory for Animal Disease Control & Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China.,Key Laboratory of Combining Farming & Animal Husbandry, Ministry of Agriculture, Animal Husbandry Research Institute, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, PR China
| | - Xin Yan
- Heilongjiang Key Laboratory for Animal Disease Control & Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Shanshan Gu
- Heilongjiang Key Laboratory for Animal Disease Control & Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Yan Yang
- Heilongjiang Key Laboratory for Animal Disease Control & Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Lili Zhao
- Heilongjiang Provincial Key Laboratory of Laboratory Animal & Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Xinmiao He
- Key Laboratory of Combining Farming & Animal Husbandry, Ministry of Agriculture, Animal Husbandry Research Institute, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, PR China
| | - Hongyan Chen
- Heilongjiang Provincial Key Laboratory of Laboratory Animal & Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Junwei Ge
- Heilongjiang Key Laboratory for Animal Disease Control & Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Di Liu
- Key Laboratory of Combining Farming & Animal Husbandry, Ministry of Agriculture, Animal Husbandry Research Institute, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, PR China
| |
Collapse
|
220
|
Staphylococcus aureus Isolated from Skin from Atopic-Dermatitis Patients Produces Staphylococcal Enterotoxin Y, Which Predominantly Induces T-Cell Receptor Vα-Specific Expansion of T Cells. Infect Immun 2020; 88:IAI.00360-19. [PMID: 31740530 DOI: 10.1128/iai.00360-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/29/2019] [Indexed: 11/20/2022] Open
Abstract
While investigating the virulence traits of Staphylococcus aureus adhering to the skin of atopic-dermatitis (AD) patients, we identified a novel open reading frame (ORF) with structural similarity to a superantigen from genome sequence data of an isolate from AD skin. Concurrently, the same ORF was identified in a bovine isolate of S. aureus and designated SElY (H. K. Ono, Y. Sato'o, K. Narita, I. Naito, et al., Appl Environ Microbiol 81:7034-7040, 2015, https://doi.org/10.1128/AEM.01873-15). Recombinant SElYbov had superantigen activity in human peripheral blood mononuclear cells. It further demonstrated emetic activity in a primate animal model, and it was proposed that SElY be renamed SEY (H. K. Ono, S. Hirose, K. Narita, M. Sugiyama, et al., PLoS Pathog 15:e1007803, 2019, https://doi.org/10.1371/journal.ppat.1007803). Here, we investigated the prevalence of the sey gene in 270 human clinical isolates of various origins in Japan. Forty-two strains were positive for the sey gene, and the positive isolates were from patients with the skin diseases atopic dermatitis and impetigo/staphylococcal scalded skin syndrome (SSSS), with a detection rate of ∼17 to 22%. There were three variants of SEY (SEY1, SEY2, and SEY3), and isolates producing SEY variants formed three distinct clusters corresponding to clonal complexes (CCs) 121, 59, and 20, respectively. Most sey + isolates produced SEY in broth culture. Unlike SEYbov, the three recombinant SEY variants exhibited stability against heat treatment. SEY predominantly activated human T cells with a particular T-cell receptor (TCR) Vα profile, a unique observation since most staphylococcal enterotoxins exert their superantigenic activities through activating T cells with specific TCR Vβ profiles. SEY may act to induce localized inflammation via skin-resident T-cell activation, facilitating the pathogenesis of S. aureus infection in disrupted epithelial barriers.
Collapse
|
221
|
Castro RD, Pedroso SHSP, Sandes SHC, Silva GO, Luiz KCM, Dias RS, Filho RAT, Figueiredo HCP, Santos SG, Nunes AC, Souza MR. Virulence factors and antimicrobial resistance of Staphylococcus aureus isolated from the production process of Minas artisanal cheese from the region of Campo das Vertentes, Brazil. J Dairy Sci 2020; 103:2098-2110. [PMID: 31980224 DOI: 10.3168/jds.2019-17138] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/28/2019] [Indexed: 12/31/2022]
Abstract
Staphylococcus aureus is one of the main pathogens found in cheeses produced with raw milk, including Minas artisanal cheese from Brazil. However, information about S. aureus isolated from artisanal cheeses and its sources of production in small-scale dairies is very limited. We aimed to characterize the virulence factors of S. aureus isolated from raw milk, endogenous starter culture, Minas artisanal cheese, and cheese handlers from the region of Campo das Vertentes, Minas Gerais, Brazil. We identified the staphylococcal isolates by MALDI-TOF mass spectrometry. We evaluated biofilm production on Congo red agar and polystyrene plates. We used PCR to detect icaA, icaB, icaC, sea, seb, sec, sed, see, tsst-1, agr, and mecA. We evaluated the expression of staphylococcal toxin genes in PCR-positive staphylococcal isolates using quantitative reverse-transcription PCR, and we evaluated the production of these toxins and their hemolytic activity in vitro. We also evaluated the antimicrobial resistance profile of the staphylococcal isolates. For statistical analysis, we used cluster analysis, χ2 tests, and correspondence tests. We analyzed 76 staphylococcal isolates. According to PCR, 18.42, 18.42, 2.63, and 77.63% were positive for sea, tsst-1, sec, and agr, respectively. We found low expression of staphylococcal toxin genes according to quantitative reverse-transcription PCR, and only 2 staphylococcal isolates produced toxic shock syndrome toxins. A total of 43 staphylococcal isolates (56.58%) had hemolytic activity; 53 were biofilm-forming on Congo red agar (69.73%), and 62 on polystyrene plates (81.58%). None of the staphylococcal isolates expressed the mecA gene, and none presented a multi-drug resistance pattern. The highest resistance was observed for penicillin G (67.11%) in 51 isolates and for tetracycline (27.63%) in 21 isolates. The staphylococcal isolates we evaluated had toxigenic potential, with a higher prevalence of sea and tsst-1. Biofilm production was the main virulence factor of the studied bacteria. Six clusters were formed whose distribution frequencies differed for hemolytic activity, biofilm formation (qualitative and quantitative analyses), and resistance to penicillin, tetracycline, and erythromycin. These findings emphasize the need for effective measures to prevent staphylococcal food poisoning by limiting S. aureus growth and enterotoxin formation throughout the food production chain and the final product.
Collapse
Affiliation(s)
- R D Castro
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - S H S P Pedroso
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - S H C Sandes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - G O Silva
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - K C M Luiz
- Laboratório de Enterotoxinas de Alimentos, Fundação Ezequiel Dias, Belo Horizonte, 30510-010, Brazil
| | - R S Dias
- Laboratório de Enterotoxinas de Alimentos, Fundação Ezequiel Dias, Belo Horizonte, 30510-010, Brazil
| | - R A T Filho
- Departamento de Engenharia de Alimentos, Universidade Federal de Viçosa, Florestal, 35690-000, Brazil
| | - H C P Figueiredo
- Departamento de Medicina Veterinária Preventiva, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - S G Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - A C Nunes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - M R Souza
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| |
Collapse
|
222
|
Epstein IJ, Rosenberg E, Stuber R, Choi MB, Donnenfeld ED, Perry HD. Double-Masked and Unmasked Prospective Study of Terpinen-4-ol Lid Scrubs With Microblepharoexfoliation for the Treatment of Demodex Blepharitis. Cornea 2020; 39:408-416. [DOI: 10.1097/ico.0000000000002243] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
223
|
Campbell AJ, Dotel R, Blyth CC, Davis JS, Tong SYC, Bowen AC. Adjunctive protein synthesis inhibitor antibiotics for toxin suppression in Staphylococcus aureus infections: a systematic appraisal. J Antimicrob Chemother 2020; 74:1-5. [PMID: 30307507 DOI: 10.1093/jac/dky387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Protein synthesis inhibitor antibiotics inhibit synthesis of new proteins, including exotoxins and other important virulence determinants in Staphylococcus aureus. A summary of the literature regarding the use of adjunctive protein synthesis inhibitors for toxin suppression in the setting of S. aureus infections is presented.
Collapse
Affiliation(s)
- A J Campbell
- Perth Children's Hospital, 15 Hospital Avenue, Perth, Western Australia, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia.,School of Medicine, Division of Paediatrics, University of Western Australia, Perth, Western Australia, Australia
| | - R Dotel
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, University of Sydney, and Department of Infectious Diseases, Blacktown Hospital, Sydney, New South Wales, Australia
| | - C C Blyth
- Perth Children's Hospital, 15 Hospital Avenue, Perth, Western Australia, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia.,School of Medicine, Division of Paediatrics, University of Western Australia, Perth, Western Australia, Australia
| | - J S Davis
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia.,Department of Infectious Diseases, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - S Y C Tong
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia.,Victorian Infectious Disease Service, The Royal Melbourne Hospital, and The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, Australia
| | - A C Bowen
- Perth Children's Hospital, 15 Hospital Avenue, Perth, Western Australia, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia.,School of Medicine, Division of Paediatrics, University of Western Australia, Perth, Western Australia, Australia.,Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| |
Collapse
|
224
|
Noli Truant S, De Marzi MC, Sarratea MB, Antonoglou MB, Meo AP, Iannantuono López LV, Fernández Lynch MJ, Todone M, Malchiodi EL, Fernández MM. egc Superantigens Impair Monocytes/Macrophages Inducing Cell Death and Inefficient Activation. Front Immunol 2020; 10:3008. [PMID: 32010128 PMCID: PMC6974467 DOI: 10.3389/fimmu.2019.03008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Bacterial superantigens (SAgs) are enterotoxins that bind to MHC-II and TCR molecules, activating as much as 20% of the T cell population and promoting a cytokine storm which enhances susceptibility to endotoxic shock, causing immunosuppression, and hindering the immune response against bacterial infection. Since monocytes/macrophages are one of the first cells SAgs find in infected host and considering the effect these cells have on directing the immune response, here, we investigated the effect of four non-classical SAgs of the staphylococcal egc operon, namely, SEG, SEI, SEO, and SEM on monocytic-macrophagic cells, in the absence of T cells. We also analyzed the molecular targets on APCs which could mediate SAg effects. We found that egc SAgs depleted the pool of innate immune effector cells and induced an inefficient activation of monocytic-macrophagic cells, driving the immune response to an impaired proinflammatory profile, which could be mediated directly or indirectly by interactions with MHC class II. In addition, performing surface plasmon resonance assays, we demonstrated that non-classical SAgs bind the gp130 molecule, which is also present in the monocytic cell surface, among other cells.
Collapse
Affiliation(s)
- Sofia Noli Truant
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mauricio C De Marzi
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina.,Instituto de Ecología y Desarrollo Sustentable (INEDES), UNLU-CONICET, Universidad Nacional de Luján, Luján, Argentina
| | - María B Sarratea
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María B Antonoglou
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana P Meo
- Hospital Dr. J. M. Ramos Mejía, Buenos Aires, Argentina
| | - Laura V Iannantuono López
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María J Fernández Lynch
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcos Todone
- Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina.,Instituto de Ecología y Desarrollo Sustentable (INEDES), UNLU-CONICET, Universidad Nacional de Luján, Luján, Argentina
| | - Emilio L Malchiodi
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa M Fernández
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
225
|
Das R, D`souza N, Choubey SK, Murlidharan S, Kurpad AV, Mandal AK. Analysis of Extracellular Proteome of Staphylococcus aureus: A Mass Spectrometry based Proteomics Method of Exotoxin Characterisation. CURR PROTEOMICS 2020. [DOI: 10.2174/1570164616666190204160627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Staphylococcus aureus (S. aureus), an important pathogen, causes a wide
range of infections in human starting from food poisoning to septicemia. It affects the host cells with
various exotoxins, known as virulence factors, which are synthesized in growth phase-dependent manner
of the bacteria. S. aureus has been reported to become resistant to antibiotics rapidly. Among two
common clinical isolates, Methicillin-sensitive S. aureus (MSSA) and Methicillin-resistant S. aureus
(MRSA), MRSA pose major problems across hospitals around the world.
Objective:
The objective of the present study was to profile the exoproteins of Methicillin-sensitive
S. aureus (ATCC 25293) and subsequently to establish a proteomics-based method of characterization
of S. aureus that is crucial in treating hospital-acquired infections.
Methods:
We used two-dimensional nanoLC/ESI-MS based proteomic platform to characterize and
quantify the exoproteins isolated from Methicillin-sensitive S. aureus (ATCC 25293) strain.
Results:
A total of 69 proteins were identified from extracellular proteome pool of ATCC 25293 strain
that includes 18 extracellular proteins, 40 cytoplasmic proteins, 2 membrane proteins, 3 cell wall proteins
and 6 uncharacterized proteins.
Conclusion:
We propose that this mass spectrometry-based proteomics method of characterization of
exoproteins might be useful to identify S. aureus strains that are resistant to antibiotics.
Collapse
Affiliation(s)
- Rajdeep Das
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Nisha D`souza
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Surya K. Choubey
- Department of Urology and Renal Transplantation, St. John’s Medical College, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Sethumadhavan Murlidharan
- Department of Microbiology, St. John’s Medical College, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Anura V. Kurpad
- Department of Physiology, St. John’s Medical College, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Amit K. Mandal
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| |
Collapse
|
226
|
Fischer AJ, Kilgore SH, Singh SB, Allen PD, Hansen AR, Limoli DH, Schlievert PM. High Prevalence of Staphylococcus aureus Enterotoxin Gene Cluster Superantigens in Cystic Fibrosis Clinical Isolates. Genes (Basel) 2019; 10:E1036. [PMID: 31842331 PMCID: PMC6947208 DOI: 10.3390/genes10121036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/04/2019] [Accepted: 12/09/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a highly prevalent respiratory pathogen in cystic fibrosis (CF). It is unclear how this organism establishes chronic infections in CF airways. We hypothesized that S. aureus isolates from patients with CF would share common virulence properties that enable chronic infection. METHODS 77 S. aureus isolates were obtained from 45 de-identified patients with CF at the University of Iowa. We assessed isolates phenotypically and used genotyping assays to determine the presence or absence of 18 superantigens (SAgs). RESULTS We observed phenotypic diversity among S. aureus isolates from patients with CF. Genotypic analysis for SAgs revealed 79.8% of CF clinical isolates carried all six members of the enterotoxin gene cluster (EGC). MRSA and MSSA isolates had similar prevalence of SAgs. We additionally observed that EGC SAgs were prevalent in S. aureus isolated from two geographically distinct CF centers. CONCLUSIONS S. aureus SAgs belonging to the EGC are highly prevalent in CF clinical isolates. The greater prevalence in these SAgs in CF airway specimens compared to skin isolates suggests that these toxins confer selective advantage in the CF airway.
Collapse
Affiliation(s)
- Anthony J. Fischer
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.B.S.); (P.D.A.); (A.R.H.)
| | - Samuel H. Kilgore
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.K.); (D.H.L.); (P.M.S.)
| | - Sachinkumar B. Singh
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.B.S.); (P.D.A.); (A.R.H.)
| | - Patrick D. Allen
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.B.S.); (P.D.A.); (A.R.H.)
| | - Alexis R. Hansen
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.B.S.); (P.D.A.); (A.R.H.)
| | - Dominique H. Limoli
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.K.); (D.H.L.); (P.M.S.)
| | - Patrick M. Schlievert
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.K.); (D.H.L.); (P.M.S.)
| |
Collapse
|
227
|
Zeng C, Liu Z, Han Z. Structure of Staphylococcal Enterotoxin N: Implications for Binding Properties to Its Cellular Proteins. Int J Mol Sci 2019; 20:ijms20235921. [PMID: 31775346 PMCID: PMC6928602 DOI: 10.3390/ijms20235921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 01/26/2023] Open
Abstract
Staphylococcus aureus strains produce a unique family of immunostimulatory exotoxins termed as bacterial superantigens (SAgs), which cross-link major histocompatibility complex class II (MHC II) molecule and T-cell receptor (TCR) to stimulate large numbers of T cells at extremely low concentrations. SAgs are associated with food poisoning and toxic shock syndrome. To date, 26 genetically distinct staphylococcal SAgs have been reported. This study reports the first X-ray structure of newly characterized staphylococcal enterotoxin N (SEN). SEN possesses the classical two domain architecture that includes an N-terminal oligonucleotide-binding fold and a C-terminal β-grasp domain. Amino acid and structure alignments revealed that several critical amino acids that are proposed to be responsible for MHC II and TCR molecule engagements are variable in SEN, suggesting that SEN may adopt a different binding mode to its cellular receptors. This work helps better understand the mechanisms of action of SAgs.
Collapse
Affiliation(s)
- Chi Zeng
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (C.Z.); (Z.L.)
- Hubei Province Fresh Food Engineering Research Center, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zhaoxin Liu
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (C.Z.); (Z.L.)
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhenggang Han
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (C.Z.); (Z.L.)
- Correspondence:
| |
Collapse
|
228
|
Tsuiji M, Shiohara K, Takei Y, Shinohara Y, Nemoto S, Yamaguchi S, Kanto M, Itoh S, Oku T, Miyashita M, Seyama Y, Kurihara M, Tsuji T. Selective Cytotoxicity of Staphylococcal α-Hemolysin (α-Toxin) against Human Leukocyte Populations. Biol Pharm Bull 2019; 42:982-988. [PMID: 31155595 DOI: 10.1248/bpb.b18-01024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Staphylococcus aureus produces a variety of exoproteins that interfere with host immune systems. We attempted to purify cytotoxins against human leukocytic cells from the culture supernatant of S. aureus by a combination of ammonium sulfate precipitation, ion-exchange chromatography on a CM-cellulose column and HPLC on a Mono S 5/50 column. A major protein possessing cytotoxicity to HL60 human promyelocytic leukemia cells was purified, and the protein was identified as α-hemolysin (Hla, α-toxin) based on its molecular weight (34 kDa) and N-terminal amino acid sequence. Flow cytometric analysis suggested differential cytotoxicity of Hla against different human peripheral blood leukocyte populations. After cell fractionation with density-gradient centrifugation, we found that peripheral blood mononuclear cells (PBMCs) were more susceptible to Hla than polymorphonuclear leukocytes. Moreover, cell surface marker analysis suggested that Hla exhibited slightly higher cytotoxicity against CD14-positive PBMCs (mainly monocytes) than CD3- or CD19-positive cells (T or B lymphocytes). From these results, we conclude that human leukocytes have different susceptibility to Hla depending on their cell lineages, and thereby the toxin may modulate the host immune response.
Collapse
Affiliation(s)
- Makoto Tsuiji
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Kazuyuki Shiohara
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Yoshinori Takei
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Yoshinori Shinohara
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Shigeyoshi Nemoto
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Satoshi Yamaguchi
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Masanori Kanto
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Saotomo Itoh
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Teruaki Oku
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Masahiro Miyashita
- Department of Clinical Chemistry, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | - Yoshiyuki Seyama
- Department of Clinical Chemistry, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| | | | - Tsutomu Tsuji
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences
| |
Collapse
|
229
|
Lahiri H, Banerjee S, Mukhopadhyay R. Free-Energy-Based Gene Mutation Detection Using LNA Probes. ACS Sens 2019; 4:2688-2696. [PMID: 31549503 DOI: 10.1021/acssensors.9b01115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We have developed a label-free approach for direct detection of gene mutations using free-energy values that are derived from single-molecule force spectroscopy (SMFS)-based nucleic acid unbinding experiments. From the duplex unbinding force values acquired by SMFS, the force-loading-rate-independent Gibbs free-energy values were derived using Jarzinsky's equality treatment. Because it provides molecule-by-molecule information, this approach is a major shift compared to the earlier reports on label-free detection of DNA sequences, which are mostly based on ensemble level data. We tested our approach in the disease model framework of multiple drug-resistant tuberculosis using the nuclease-resistant and conformationally rigid locked nucleic acid probes that are a robust and efficient alternative to the DNA probes. All of the major mutations in Mycobacterium tuberculosis (MTB), as relevant to MTB's resistance to the first-line anti-TB drugs rifampicin and isoniazid, could be identified, and the wild type could be discriminated from the most prevalent mutation and the most prevalent mutation from the less occurring ones. Our approach could also identify DNA sequences (45 mer), having overhang stretches at different positions with respect to the complementary stretch. Probably for the first time, the findings show that free-energy-based detection of gene mutations is possible at molecular resolution.
Collapse
Affiliation(s)
- Hiya Lahiri
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Siddhartha Banerjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Rupa Mukhopadhyay
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| |
Collapse
|
230
|
Abstract
The complex regulatory role of the proteases necessitates very tight coordination and control of their expression. While this process has been well studied, a major oversight has been the consideration of proteases as a single entity rather than as 10 enzymes produced from four different promoters. As such, in this study, we comprehensively characterized the regulation of each protease promoter, discovering vast differences in the way each protease operon is controlled. Additionally, we broaden the picture of protease regulation using a global screen to identify novel loci controlling protease activity, uncovering a cadre of new effectors of protease expression. The impact of these elements on the activity of proteases and known regulators was characterized by producing a comprehensive regulatory circuit that emphasizes the complexity of protease regulation in Staphylococcus aureus. A primary function of the extracellular proteases of Staphylococcus aureus is to control the progression of infection by selectively modulating the stability of virulence factors. Consequently, a regulatory network exists to titrate protease abundance/activity to influence the accumulation, or lack thereof, of individual virulence factors. Herein, we comprehensively map this system, exploring the regulation of the four protease loci by known and novel factors. In so doing, we determined that seven major elements (SarS, SarR, Rot, MgrA, CodY, SaeR, and SarA) form the primary network of control, with the latter three being the most powerful. We note that expression of aureolysin is largely repressed by these factors, while the spl operon is subject to the strongest upregulation of any protease loci, particularly by SarR and SaeR. Furthermore, when exploring scpA expression, we find it to be profoundly influenced in opposing fashions by SarA (repressor) and SarR (activator). We also present the screening of >100 regulator mutants of S. aureus, identifying 7 additional factors (ArgR2, AtlR, MntR, Rex, XdrA, Rbf, and SarU) that form a secondary circuit of protease control. Primarily, these elements serve as activators, although we reveal XdrA as a new repressor of protease expression. With the exception or ArgR2, each of the new effectors appears to work through the primary network of regulation to influence protease production. Collectively, we present a comprehensive regulatory circuit that emphasizes the complexity of protease regulation and suggest that its existence speaks to the importance of these enzymes to S. aureus physiology and pathogenic potential. IMPORTANCE The complex regulatory role of the proteases necessitates very tight coordination and control of their expression. While this process has been well studied, a major oversight has been the consideration of proteases as a single entity rather than as 10 enzymes produced from four different promoters. As such, in this study, we comprehensively characterized the regulation of each protease promoter, discovering vast differences in the way each protease operon is controlled. Additionally, we broaden the picture of protease regulation using a global screen to identify novel loci controlling protease activity, uncovering a cadre of new effectors of protease expression. The impact of these elements on the activity of proteases and known regulators was characterized by producing a comprehensive regulatory circuit that emphasizes the complexity of protease regulation in Staphylococcus aureus.
Collapse
|
231
|
Toxic Shock Syndrome Toxin 1-Producing Methicillin-Resistant Staphylococcus aureus of Clonal Complex 5, the New York/Japan Epidemic Clone, Causing a High Early-Mortality Rate in Patients with Bloodstream Infections. Antimicrob Agents Chemother 2019; 63:AAC.01362-19. [PMID: 31501145 DOI: 10.1128/aac.01362-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/30/2019] [Indexed: 12/23/2022] Open
Abstract
This study was performed to evaluate the clinical impacts of putative risk factors in patients with Staphylococcus aureus bloodstream infections (BSIs) through a prospective, multicenter, observational study. All 567 patients with S. aureus BSIs that occurred during a 1-year period in six general hospitals were included in this study. Host- and pathogen-related variables were investigated to determine risk factors for the early mortality of patients with S. aureus BSIs. The all-cause mortality rate was 15.0% (85/567) during the 4-week follow-up period from the initial blood culture, and 76.5% (65/85) of the mortality cases occurred within the first 2 weeks. One-quarter (26.8%, 152/567) of the S. aureus blood isolates carried the tst-1 gene, and most (86.2%, 131/152) of them were identified to be clonal complex 5 agr type 2 methicillin-resistant S. aureus (MRSA) strains harboring staphylococcal cassette chromosome mec type II, belonging to the New York/Japan epidemic clone. A multivariable logistic regression showed that the tst-1 positivity of the causative S. aureus isolates was associated with an increased 2-week mortality rate both in patients with S. aureus BSIs (adjusted odds ratio [aOR], 1.62; 95% confidence interval [CI], 0.90 to 2.88) and in patients with MRSA BSIs (aOR, 2.61; 95% CI, 1.19 to 6.03). Two host-related factors, an increased Pitt bacteremia score and advanced age, as well as a pathogen-related factor, carriage of tst-1 by causative MRSA isolates, were risk factors for 2-week mortality in patients with BSIs. Careful management of patients with BSIs caused by the New York/Japan epidemic clone is needed to improve clinical outcomes.
Collapse
|
232
|
Zheng Y, Qin C, Zhang X, Zhu Y, Li A, Wang M, Tang Y, Kreiswirth BN, Chen L, Zhang H, Du H. The tst gene associated Staphylococcus aureus pathogenicity island facilitates its pathogenesis by promoting the secretion of inflammatory cytokines and inducing immune suppression. Microb Pathog 2019; 138:103797. [PMID: 31614194 DOI: 10.1016/j.micpath.2019.103797] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 11/25/2022]
Abstract
Staphylococcus aureus (S. aureus) is an important pathogen causing various limited or systemic infections. Methicillin resistant S. aureus (MRSA) in particular presents a major clinical and public health problem. Toxic shock syndrome toxin-1 (TSST-1) encoded by the gene tst is an important virulence factor of tst positive S. aureus, leading to multi-organ malfunction. However, the mechanism of TSST-1 in pathogenesis is only partly clear. In this study, we investigated the prevalence of the tst gene in clinical isolates of S. aureus. Then, animal experiments were performed to further evaluate the influence of the presence of the tst gene associated Staphylococcus aureus Pathogenicity Island (SaPI) on body weight, serum cytokine concentrations and the bacterial load in different organs. In addition, macrophages were used to analyze the secretion of cytokines in vitro and bacterial survival in the cytoplasm. Finally, pathological analysis was carried out to evaluate organ tissue impairment. The results demonstrated that the prevalence of tst gene was approximately 17.8% of the bacterial strains examined. BALB/c mice infected with tst gene associated SaPI positive isolates exhibited a severe loss of body weight and a high bacterial load in the liver, heart, kidney and spleen. Pathological analysis demonstrated that tissue impairment was more severe after infection with tst gene associated SaPI positive isolates. Moreover, the secretion of IL-6, IL-2 and IL17A by macrophages infected with tst gene associated SaPI positive isolates clearly increased. Notably, IL-6 secretion in BALB/c mice infected with tst gene associated SaPI positive isolates was higher than that in BALB/c mice infected with negative ones. Together, these results indicated that the tst gene associated SaPI may play a critical role in the pathological process of infection via a direct and persistent toxic function, and by promoting the secretion of inflammatory cytokines that indirectly induce immune suppression.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China
| | - Chenhao Qin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China
| | - Xianfeng Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China
| | - Yifan Zhu
- School of Psychiatry, North Sichuan Medical College, Nanchong, Sichuan, 637007, PR China
| | - Aiqing Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China
| | - Yiwei Tang
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Barry N Kreiswirth
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Liang Chen
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, PR China.
| |
Collapse
|
233
|
Labruère R, Sona AJ, Turos E. Anti-Methicillin-Resistant Staphylococcus aureus Nanoantibiotics. Front Pharmacol 2019; 10:1121. [PMID: 31636560 PMCID: PMC6787278 DOI: 10.3389/fphar.2019.01121] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/30/2019] [Indexed: 01/08/2023] Open
Abstract
Nanoparticle-based antibiotic constructs have become a popular area of investigation in the biomedical sciences. Much of this work has pertained to human diseases, largely in the cancer therapy arena. However, considerable research has also been devoted to the nanochemistry for controlling infectious diseases. Among these are ones due to bacterial infections, which can cause serious illnesses leading to death. The onset of multi-drug-resistant (MDR) infections such as those caused by the human pathogen Staphylococcus aureus has created a dearth of problems such as surgical complications, persistent infections, and lack of available treatments. In this article, we set out to review the primary literature on the design and development of new nanoparticle materials for the potential treatment of S. aureus infections, and areas that could be further expanded upon to make nanoparticle antibiotics a mainstay in clinical settings.
Collapse
Affiliation(s)
- Raphaël Labruère
- Institut de Chimie Moléculaire et des Matériaux d’Orsay (ICMMO), CNRS, Univ Paris Sud, Université Paris-Saclay, Orsay, France
| | - A. J. Sona
- Center for Molecular Diversity in Drug Design, Discovery and Delivery, Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Edward Turos
- Center for Molecular Diversity in Drug Design, Discovery and Delivery, Department of Chemistry, University of South Florida, Tampa, FL, United States
| |
Collapse
|
234
|
Li W, Geng X, Liu D, Li Z. Near-Infrared Light-Enhanced Protease-Conjugated Gold Nanorods As A Photothermal Antimicrobial Agent For Elimination Of Exotoxin And Biofilms. Int J Nanomedicine 2019; 14:8047-8058. [PMID: 31632017 PMCID: PMC6781946 DOI: 10.2147/ijn.s212750] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022] Open
Abstract
Purpose Treatment strategies to eliminate bacterial infections have long emphasized bacterial killing as a goal. However, bacteria secrete toxins that sustain chronic disease and dead cells release DNA that can promote the spread of antibiotic resistance even when viable cells are eradicated. Meanwhile, biofilms regulated by quorum-sensing system, protect bacteria and promote the development of antibiotic resistance. Thus, all of these factors underscore the need for novel antimicrobial therapeutic treatments as alternatives to traditional antibiotics. Here, a smart material was developed that incorporated gold nanorods and an adsorbed protease (protease-conjugated gold nanorods, PGs). When illuminated with near-infrared (NIR) light, PGs functioned to physically damage bacteria, prevent biofilm and exotoxin production, eliminate pre-existing biofilm and exotoxin, and inhibit bacterial quorum-sensing systems. Methods PGs were incubated with suspensions of Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria followed by exposure to 808-nm NIR laser irradiation. Bacterial viability was determined using a colony-forming unit assay followed by an exploration of cell-damage mechanisms using transmission electron microscopy, scanning electron microscopy, agarose gel electrophoresis, and SDS-PAGE. Quantification of biofilm mass was performed using crystal violet staining. A commercial enterotoxin ELISA kit was used to test inhibitory and degradative effects of PGs on secreted exotoxin. Results Use of the remote-controlled antibacterial system reduced surviving bacterial populations to 3.2% and 2.1% of untreated control numbers for E. coli and S. aureus, respectively, and inhibited biofilm formation and exotoxin secretion even in the absence of NIR radiation. However, enhanced degradation of existing biofilm and exotoxin was observed when PGs were used with NIR laser irradiation. Conclusion This promising new strategy achieved both the reduction of viable microorganisms and elimination of biofilm and exotoxin. Thus, this strategy addresses the long-ignored issue of persistence of bacterial residues that perpetuate chronic illness in patients even after viable bacteria have been eradicated.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China.,Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, People's Republic of China
| | - Xu Geng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China
| | - Dongni Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China
| | - Zhengqiang Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China
| |
Collapse
|
235
|
Amoako DG, Somboro AM, Abia ALK, Allam M, Ismail A, Bester LA, Essack SY. Genome Mining and Comparative Pathogenomic Analysis of An Endemic Methicillin-Resistant Staphylococcus Aureus (MRSA) Clone, ST612-CC8-t1257-SCCmec_IVd(2B), Isolated in South Africa. Pathogens 2019; 8:E166. [PMID: 31569754 PMCID: PMC6963616 DOI: 10.3390/pathogens8040166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
This study undertook genome mining and comparative genomics to gain genetic insights into the dominance of the methicillin-resistant Staphylococcus aureus (MRSA) endemic clone ST612-CC8-t1257-SCCmec_IVd(2B), obtained from the poultry food chain in South Africa. Functional annotation of the genome revealed a vast array of similar central metabolic, cellular and biochemical networks within the endemic clone crucial for its survival in the microbial community. In-silico analysis of the clone revealed the possession of uniform defense systems, restriction-modification system (type I and IV), accessory gene regulator (type I), arginine catabolic mobile element (type II), and type 1 clustered, regularly interspaced, short palindromic repeat (CRISPR)Cas array (N = 7 ± 1), which offer protection against exogenous attacks. The estimated pathogenic potential predicted a higher probability (average Pscore ≈ 0.927) of the clone being pathogenic to its host. The clone carried a battery of putative virulence determinants whose expression are critical for establishing infection. However, there was a slight difference in their possession of adherence factors (biofilm operon system) and toxins (hemolysins and enterotoxins). Further analysis revealed a conserved environmental tolerance and persistence mechanisms related to stress (oxidative and osmotic), heat shock, sporulation, bacteriocins, and detoxification, which enable it to withstand lethal threats and contribute to its success in diverse ecological niches. Phylogenomic analysis with close sister lineages revealed that the clone was closely related to the MRSA isolate SHV713 from Australia. The results of this bioinformatic analysis provide valuable insights into the biology of this endemic clone.
Collapse
Affiliation(s)
- Daniel Gyamfi Amoako
- Infection Genomics and Applied Bioinformatics Division, Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
- Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal; Durban 4000, South Africa.
| | - Anou M Somboro
- Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal; Durban 4000, South Africa.
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Akebe Luther King Abia
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Mushal Allam
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg 2131, South Africa.
| | - Arshad Ismail
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg 2131, South Africa.
| | - Linda A Bester
- Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal; Durban 4000, South Africa.
| | - Sabiha Y Essack
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| |
Collapse
|
236
|
Staphylococcus aureus enterotoxin genes detected in milk from various livestock species in northern pastoral region of Kenya. Food Control 2019. [DOI: 10.1016/j.foodcont.2019.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
237
|
Guo N, Liu Z, Yan Z, Liu Z, Hao K, Liu C, Wang J. Subinhibitory concentrations of Honokiol reduce α-Hemolysin (Hla) secretion by Staphylococcus aureus and the Hla-induced inflammatory response by inactivating the NLRP3 inflammasome. Emerg Microbes Infect 2019; 8:707-716. [PMID: 31119985 PMCID: PMC6534259 DOI: 10.1080/22221751.2019.1617643] [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] [Indexed: 01/02/2023]
Abstract
Staphylococcus aureus (S. aureus) is one of the most serious human pathogens. α-Hemolysin (Hla) secreted by S. aureus is a key toxin for various infections. We herein report that Honokiol, a natural plant polyphenol, inhibits the secretion and hemolytic activity of staphylococcal Hla with concomitant growth inhibition of S. aureus and protection of S. aureus-mediated cell injury within subinhibitory concentrations. In parallel, Honokiol attenuates the staphylococcal Hla-induced inflammatory response by inhibiting NLRP3 inflammasome activation in vitro and in vivo. Consequently, the biologically active forms of the inflammatory cytokines IL-1β and IL-18 are reduced significantly in response to Honokiol in mice infected with S. aureus. Experimentally, we confirm that Honokiol binds to monomeric Hla with a modest affinity without impairing its oligomerization. Based on molecular docking analyses in silico, we make a theoretical discovery that Honokiol is located outside of the triangular region of monomeric Hla. The binding model restricts the function of the residues related to membrane channel formation, which leads to the functional disruption of the assembled membrane channel. This research creates a new paradigm for developing therapeutic agents against staphylococcal Hla-mediated infections.
Collapse
Affiliation(s)
- Na Guo
- a State Key Laboratory of Electroanalytical Chemistry , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun , People's Republic of China.,c Department of Food Quality and Safety , College of Food Science and Engineering, Jilin University , Changchun , People's Republic of China
| | - Zuojia Liu
- a State Key Laboratory of Electroanalytical Chemistry , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun , People's Republic of China
| | - Zhiqiang Yan
- a State Key Laboratory of Electroanalytical Chemistry , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun , People's Republic of China
| | - Zonghui Liu
- c Department of Food Quality and Safety , College of Food Science and Engineering, Jilin University , Changchun , People's Republic of China
| | - Kun Hao
- c Department of Food Quality and Safety , College of Food Science and Engineering, Jilin University , Changchun , People's Republic of China
| | - Chuanbo Liu
- a State Key Laboratory of Electroanalytical Chemistry , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun , People's Republic of China
| | - Jin Wang
- a State Key Laboratory of Electroanalytical Chemistry , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun , People's Republic of China.,b Department of Chemistry and Physics , State University of New York , Stony Brook , NY , USA
| |
Collapse
|
238
|
Barie PS, Narayan M, Sawyer RG. Immunization Against Staphylococcus aureus Infections. Surg Infect (Larchmt) 2019; 19:750-756. [PMID: 31033407 DOI: 10.1089/sur.2018.263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Infections caused by Staphylococcus aureus continue to plague surgical patients, whether as surgical site infections or other nosocomial infections that complicate surgical care. The only meaningful methods available to decrease the risk of developing such infections are topical skin antisepsis (pre-operative skin preparation) and peri-operative antibiotic prophylaxis, neither of which offer a panacea. Alternatives to the latter are sought so as to minimize antibiotic selection pressure as a factor in the increasing problem of antimicrobial drug resistance. This review considers the possibility that immunization against S. aureus may offer a viable alternative for prophylaxis. Methods: Review and synthesis of pertinent English-language medical literature. Results: Vaccination against viral pathogens has been in successful clinical use for more than two centuries and was instrumental in the eradication of smallpox and the near-elimination of diseases such as poliomyelitis. Vaccinations against a limited number of bacterial pathogens (e.g., Bordetella pertussis, Clostridium tetanii, Corynebacterium diphtheriae, Haemophilus influenzae type b, Neisseria meningiditis, Streptococcus pneumoniae) have also been introduced with success, whereas others against bacteria are in development (C. difficile, Pseudomonas aeruginosa, S. aureus). Vaccination against S. aureus infection is in current veterinary use (e.g., to prevent mastitis among dairy cattle) but has not been successful to date in human beings despite multiple attempts, although development continues. Conclusions: Because of its complex microbiology, including multiple virulence factors and the ability to evade host immune surveillance, S. aureus presents numerous antigenic targets for vaccine development. Failure of two prior single-antigen vaccines in clinical trials has led to the consensus that future vaccine candidates must be directed against multiple antigens. Two distinct four-antigen vaccines are in clinical trials, but efficacy is yet to be determined.
Collapse
Affiliation(s)
- Philip S Barie
- 1 Department of Surgery, Weill Cornell Medicine , New York, New York
- 2 Department of Medicine, Weill Cornell Medicine , New York, New York
| | - Mayur Narayan
- 1 Department of Surgery, Weill Cornell Medicine , New York, New York
| | - Robert G Sawyer
- 3 Department of Surgery, Western Michigan University , Kalamazoo, Michigan
| |
Collapse
|
239
|
Schwan WR. Staphylococcus aureus Toxins: Armaments for a Significant Pathogen. Toxins (Basel) 2019; 11:toxins11080457. [PMID: 31382602 PMCID: PMC6724065 DOI: 10.3390/toxins11080457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus species are common inhabitants of humans and other animals [...].
Collapse
Affiliation(s)
- William R Schwan
- Department of Microbiology, University of Wisconsin-La Crosse, 1725 State St., La Crosse, WI 54601, USA.
| |
Collapse
|
240
|
Kobayashi T, Nakaminami H, Ohtani H, Yamada K, Nasu Y, Takadama S, Noguchi N, Fujii T, Matsumoto T. An outbreak of severe infectious diseases caused by methicillin-resistant Staphylococcus aureus USA300 clone among hospitalized patients and nursing staff in a tertiary care university hospital. J Infect Chemother 2019; 26:76-81. [PMID: 31375457 DOI: 10.1016/j.jiac.2019.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 11/30/2022]
Abstract
The USA300 clone, which produces Panton-Valentine leukocidin (PVL), is a major highly pathogenic community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) clone that is spreading throughout the world. Although the prevalence of the USA300 clone in Japan was very limited a decade ago, its incidence has been increasing in both community and hospital settings in recent years. There is great concern that the USA300 clone will cause more complicated diseases and become a serious threat to immunocompromised patients in hospital settings. Here, we report an outbreak of severe infectious diseases in a tertiary care university hospital involving the incidence of deep infections, including bacteremia, and continuous and frequent isolation of MRSA strains for five months from six patients and a healthy nursing staff member in the same ward. The genotype of all MRSA isolates was identical to that of the USA300 clone. Furthermore, pulsed-field gel electrophoresis analysis indicated that all MRSA had the same patterns. These data demonstrate that a USA300 clone outbreak had occurred in the hospital. Fortunately, this outbreak was terminated subsequent to the interventions of the infection control team and all patients recovered following the appropriate therapies. Our report demonstrates that patients carrying highly pathogenic CA-MRSA have the potential to become a source of nosocomial outbreaks that can spread to healthy healthcare workers. Therefore, stricter standard precautions should be applied for all patients at the time of admission to prevent such nosocomial outbreaks.
Collapse
Affiliation(s)
- Takehito Kobayashi
- Department of Infection Prevention and Control, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; Department of Microbiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Hidemasa Nakaminami
- Department of Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hiroshi Ohtani
- Division of General Medicine, Tachikawa Sogo Hospital, 4-1 Midori-Cho, Tachikawa, Tokyo 190-8578, Japan
| | - Kanako Yamada
- Department of Infection Prevention and Control, Tokyo Medical University Hachioji Medical Center, 1163 Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Yutaka Nasu
- Department of Infection Prevention and Control, Tokyo Medical University Hachioji Medical Center, 1163 Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Shunsuke Takadama
- Department of Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Norihisa Noguchi
- Department of Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Takeshi Fujii
- Department of Infection Prevention and Control, Tokyo Medical University Hachioji Medical Center, 1163 Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Tetsuya Matsumoto
- Department of Microbiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; Department of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita City, Chiba Prefecture, 286-8686, Japan.
| |
Collapse
|
241
|
Hietala V, Horsma-Heikkinen J, Carron A, Skurnik M, Kiljunen S. The Removal of Endo- and Enterotoxins From Bacteriophage Preparations. Front Microbiol 2019; 10:1674. [PMID: 31396188 PMCID: PMC6664067 DOI: 10.3389/fmicb.2019.01674] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
The production of phages for therapeutic purposes demands fast, efficient and scalable purification procedures. Phage lysates have a wide range of impurities, of which endotoxins of gram-negative bacteria and protein toxins produced by many pathogenic bacterial species are harmful to humans. The highest allowed endotoxin concentration for parenterally applied medicines is 5 EU/kg/h. The aim of this study was to evaluate the feasibility of different purification methods in endotoxin and protein toxin removal in the production of phage preparations for clinical use. In the purification assays, we utilized three phages: Escherichia phage vB_EcoM_fHoEco02, Acinetobacter phage vB_ApiM_fHyAci03, and Staphylococcus phage vB_SauM_fRuSau02. The purification methods tested in the study were precipitation with polyethylene glycol, ultracentrifugation, ultrafiltration, anion exchange chromatography, octanol extraction, two different endotoxin removal columns, and different combinations thereof. The efficiency of the applied purification protocols was evaluated by measuring phage titer and either endotoxins or staphylococcal enterotoxins A and C (SEA and SEC, respectively) from samples taken from different purification steps. The most efficient procedure in endotoxin removal was the combination of ultrafiltration and EndoTrap HD affinity column, which was able to reduce the endotoxin-to-phage ratio of vB_EcoM_fHoEco02 lysate from 3.5 × 104 Endotoxin Units (EU)/109 plaque forming units (PFU) to 0.09 EU/109 PFU. The combination of ultrafiltration and anion exchange chromatography resulted in ratio 96 EU/109 PFU, and the addition of octanol extraction step into this procedure still reduced this ratio threefold. The other methods tested either resulted to less efficient endotoxin removal or required the use of harmful chemicals that should be avoided when producing phage preparations for medical use. Ultrafiltration with 100,000 MWCO efficiently removed enterotoxins from vB_SauM_fRuSau02 lysate (from 1.3 to 0.06 ng SEA/109 PFU), and anion exchange chromatography reduced the enterotoxin concentration below 0.25 ng/ml, the detection limit of the assay.
Collapse
Affiliation(s)
- Ville Hietala
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Horsma-Heikkinen
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Division of Clinical Microbiology, HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Annelie Carron
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Division of Clinical Microbiology, HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Saija Kiljunen
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
242
|
Sharma-Kuinkel BK, Tkaczyk C, Bonnell J, Yu L, Tovchigrechko A, Tabor DE, Park LP, Ruffin F, Esser MT, Sellman BR, Fowler VG, Ruzin A. Associations of pathogen-specific and host-specific characteristics with disease outcome in patients with Staphylococcus aureus bacteremic pneumonia. Clin Transl Immunology 2019; 8:e01070. [PMID: 31360464 PMCID: PMC6640002 DOI: 10.1002/cti2.1070] [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] [Received: 03/06/2019] [Revised: 06/24/2019] [Accepted: 06/30/2019] [Indexed: 11/08/2022] Open
Abstract
Objective To understand the relationships of Staphylococcus aureus (SA) bacteremic pneumonia (SABP) outcome with patient‐specific and SA‐specific variables. Methods We analysed SA bloodstream isolates and matching sera in SABP patients by sequencing SA isolates (n = 50) and measuring in vitro AT production, haemolytic activity and expression of ClfA and ClfB. Controls were sera from gram‐negative bacteremia patients with or without pneumonia and uninfected subjects. Levels of IgGs, IgMs and neutralizing antibodies (NAbs) against SA antigens were quantified and analysed by one‐way ANOVA. Associations of patient outcomes with patient variables, antibody levels and isolate characteristics were evaluated by univariate and multivariate logistic regression analyses. Results SABP patients had higher levels of IgGs against eight virulence factors and anti‐alpha toxin (AT) NAbs than uninfected controls. Levels of IgG against AT and IgMs against ClfA, FnbpA and SdrC were higher in clinically cured SABP patients than in clinical failures. Anti‐LukAB NAb levels were elevated in all cohorts. Increased odds of cure correlated with higher haemolytic activity of SA strains, longer time between surgery and bacteremia (> 30 days), longer duration of antibiotic therapy, lower acute physiology and total APACHE II scores, lack of persistent fever for > 72 h and higher levels of antibodies against AT (IgG), ClfA (IgM), FnbpA (IgM) and SdrC (IgM). Discussion Limitations included the cross‐sectional observational nature of the study, small sample size and inability to measure antibody levels against all SA virulence factors. Conclusion Our results suggest that SABP patients may benefit from immunotherapy targeting multiple SA antigens.
Collapse
Affiliation(s)
- Batu K Sharma-Kuinkel
- Division of Infectious Diseases, Department of Medicine, Duke University Durham NC USA
| | | | | | - Li Yu
- Statistical Sciences, AstraZeneca Gaithersburg MD USA
| | | | | | - Lawrence P Park
- Division of Infectious Diseases, Department of Medicine, Duke University Durham NC USA
| | - Felicia Ruffin
- Division of Infectious Diseases, Department of Medicine, Duke University Durham NC USA
| | - Mark T Esser
- Microbial Sciences, AstraZeneca Gaithersburg MD USA
| | | | - Vance G Fowler
- Division of Infectious Diseases, Department of Medicine, Duke University Durham NC USA
| | - Alexey Ruzin
- Microbial Sciences, AstraZeneca Gaithersburg MD USA
| |
Collapse
|
243
|
Staphylococcus aureus Toxins: From Their Pathogenic Roles to Anti-virulence Therapy Using Natural Products. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0059-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
244
|
Regulation of the Staphylococcal Superantigen-Like Protein 1 Gene of Community-Associated Methicillin-Resistant Staphylococcus aureus in Murine Abscesses. Toxins (Basel) 2019; 11:toxins11070391. [PMID: 31277443 PMCID: PMC6669464 DOI: 10.3390/toxins11070391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022] Open
Abstract
Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) causes substantial skin and soft tissue infections annually in the United States and expresses numerous virulence factors, including a family of toxins known as the staphylococcal superantigen-like (SSL) proteins. Many of the SSL protein structures have been determined and implicated in immune system avoidance, but the full scope that these proteins play in different infection contexts remains unknown and continues to warrant investigation. Analysis of ssl gene regulation may provide valuable information related to the function of these proteins. To determine the transcriptional regulation of the ssl1 gene of CA-MRSA strain MW2, an ssl1 promoter::lux fusion was constructed and transformed into S.aureus strains RN6390 and Newman. Resulting strains were grown in a defined minimal medium (DSM) broth and nutrient-rich brain-heart infusion (BHI) broth and expression was determined by luminescence. Transcription of ssl1 was up-regulated and occurred earlier during growth in DSM broth compared to BHI broth suggesting expression is regulated by nutrient availability. RN6390 and Newman strains containing the ssl1::lux fusion were also used to analyze regulation in vivo using a mouse abscess model of infection. A marked increase in ssl1 transcription occurred early during infection, suggesting SSL1 is important during early stages of infection, perhaps to avoid the immune system.
Collapse
|
245
|
Rezaeinasab M, Benvidi A, Gharaghani S, Zare HR. Chemometrics approaches based on electrochemical methods for the investigation of interaction between bovine serum albumin and carvacrol with the aim of its application to protein sensing. J Electroanal Chem (Lausanne) 2019. [DOI: 10.1016/j.jelechem.2019.05.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
246
|
Kannappan A, Srinivasan R, Nivetha A, Annapoorani A, Pandian SK, Ravi AV. Anti-virulence potential of 2-hydroxy-4-methoxybenzaldehyde against methicillin-resistant Staphylococcus aureus and its clinical isolates. Appl Microbiol Biotechnol 2019; 103:6747-6758. [PMID: 31230099 DOI: 10.1007/s00253-019-09941-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 05/09/2019] [Accepted: 05/25/2019] [Indexed: 12/28/2022]
Abstract
Burgeoning antibiotic resistance among bacterial pathogens necessitates the alternative treatment options to control the multidrug-resistant bacterial infections. Plant secondary metabolites, a significant source of structurally diverse compounds, posses several biological activities. The present study was designed to investigate the anti-virulence potential of least explored phytocompound 2-hydroxy-4-methoxybenzaldehyde (HMB) against methicillin-resistant Staphylococcus aureus (MRSA) and its clinical isolates. The minimum inhibitory concentration of HMB was found to be 1024 μg/ml. HMB at sub-MIC (200 μg/ml) exhibited a profound staphyloxanthin inhibitory activity against MRSA and its clinical isolates. Besides, growth curve analysis revealed the non-bactericidal activity of HMB at its sub-MIC. Other virulences of MRSA such as lipase, nuclease, and hemolysin were also significantly inhibited upon HMB treatment. The observations made out of blood and H2O2 sensitivity assay suggested that HMB treatment sensitized the test pathogens and aided the functions of host immune responses. Transcriptomic analysis revealed that HMB targets the virulence regulatory genes such as sigB and saeS to attenuate the production of virulence arsenal in MRSA. Further, the result of in vitro cytotoxicity assay using PBMC cells portrayed the non-toxic nature of HMB. To our knowledge, for the first time, the present study reported the virulence inhibitory property of HMB against MRSA along with plausible molecular mechanisms. Additional studies incorporating in vivo analysis and omics technologies are required to explore the anti-virulence potential of HMB and its mode of action during MRSA infections.
Collapse
Affiliation(s)
- Arunachalam Kannappan
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | - Ramanathan Srinivasan
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | - Arumugam Nivetha
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | - Angusamy Annapoorani
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | | | - Arumugam Veera Ravi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, 630003, India.
| |
Collapse
|
247
|
Dotel R, Tong SYC, Bowen A, Nelson JN, O’Sullivan MVN, Campbell AJ, McMullan BJ, Britton PN, Francis JR, Eisen DP, Robinson O, Manning L, Davis JS. CASSETTE-clindamycin adjunctive therapy for severe Staphylococcus aureus treatment evaluation: study protocol for a randomised controlled trial. Trials 2019; 20:353. [PMID: 31196132 PMCID: PMC6567404 DOI: 10.1186/s13063-019-3452-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Exotoxins are important virulence factors in Staphylococcus aureus. Clindamycin, a protein synthesis inhibitor antibiotic, is thought to limit exotoxin production and improve outcomes in severe S. aureus infections. However, randomised prospective data to support this are lacking. METHODS An open-label, multicentre, randomised controlled trial (RCT) will compare outcome differences in severe S. aureus infection between standard treatment (flucloxacillin/cefazolin in methicillin-susceptible S. aureus; and vancomycin/daptomycin in methicillin-resistant S. aureus) and standard treatment plus an additional clindamycin given for 7 days. We will include a minimum of 60 participants (both adult and children) in the pilot study. Participants will be enrolled within 72 h of an index culture. Severe infections will include septic shock, necrotising pneumonia, or multifocal and non-contiguous skin and soft tissue/osteoarticular infections. Individuals who are immunosuppressed, moribund, with current severe diarrhoea or Clostridiodes difficile infection, pregnant, and those with anaphylaxis to β-lactams or lincosamides will be excluded. The primary outcomes measure is the number of days alive and free (1 or 0) of systemic inflammatory response syndrome (SIRS) within the first 14 days post randomisation. The secondary outcomes measure will include all-cause mortality at 14, 42, and 90 days, time to resolution of SIRS, proportion with microbiological treatment failure, and rate of change of C-reactive protein over time. Impacts of inducible clindamycin resistance, strain types, methicillin susceptibility, and presence of various exotoxins will also be analysed. DISCUSSION This study will assess the effect of adjunctive clindamycin on patient-centred outcomes in severe, toxin-mediated S. aureus infections. The pilot study will provide feasibility for a much larger RCT. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry, ACTRN12617001416381p . Registered on 6 October 2017.
Collapse
Affiliation(s)
- Ravindra Dotel
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, University of Sydney, Sydney, Australia
- Department of Infectious Diseases, Blacktown Hospital, 18 Blacktown Road, Blacktown, NSW 2148 Australia
| | - Steven Y. C. Tong
- Victorian Infectious Disease Service, The Royal Melbourne Hospital–Doherty Department, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Menzies School of Health Research, Darwin, Australia
| | - Asha Bowen
- Department of Infectious Diseases, Perth Children’s Hospital, Perth, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | | | - Matthew V. N. O’Sullivan
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, University of Sydney, Sydney, Australia
- New South Wales Health Pathology, Newcastle, Australia
| | - Anita J. Campbell
- Department of Infectious Diseases, Perth Children’s Hospital, Perth, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Brendan J. McMullan
- Department of Immunology and Infectious Diseases, Sydney Children’s Hospital, Sydney, Australia
- School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
- National Centre for Infections in Cancer, University of Melbourne, Melbourne, Australia
| | - Philip N. Britton
- Department of Infectious Diseases and Microbiology, Children’s Hospital Westmead, Sydney, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School and Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Joshua R. Francis
- Menzies School of Health Research, Darwin, Australia
- Department of Paediatrics, Royal Darwin Hospital, Darwin, Australia
| | - Damon P. Eisen
- Townsville Hospital and Health Service, Townsville, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Owen Robinson
- Department of Infectious Diseases, Royal Perth Hospital, Perth, Australia
- Department of Infectious Diseases, Fiona Stanley Hospital, Perth, Australia
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, WA Australia
- Antimicrobial Resistance and Infectious Diseases Research Laboratory, School of Veterinary and Life Sciences, Murdoch University, Perth, WA Australia
| | - Laurens Manning
- Department of Infectious Diseases, Fiona Stanley Hospital, Perth, Australia
- Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Joshua S. Davis
- Menzies School of Health Research, Darwin, Australia
- John Hunter Hospital, University of Newcastle, Newcastle, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| |
Collapse
|
248
|
Zhao X, Palma Medina LM, Stobernack T, Glasner C, de Jong A, Utari P, Setroikromo R, Quax WJ, Otto A, Becher D, Buist G, van Dijl JM. Exoproteome Heterogeneity among Closely Related Staphylococcus aureus t437 Isolates and Possible Implications for Virulence. J Proteome Res 2019; 18:2859-2874. [PMID: 31119940 PMCID: PMC6617432 DOI: 10.1021/acs.jproteome.9b00179] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus with spa-type t437 has been identified as a predominant community-associated methicillin-resistant S. aureus clone from Asia, which is also encountered in Europe. Molecular typing has previously shown that t437 isolates are highly similar regardless of geographical regions or host environments. The present study was aimed at assessing to what extent this high similarity is actually reflected in the production of secreted virulence factors. We therefore profiled the extracellular proteome, representing the main reservoir of virulence factors, of 20 representative clinical isolates by mass spectrometry. The results show that these isolates can be divided into three groups and nine subgroups based on exoproteome abundance signatures. This implies that S. aureus t437 isolates show substantial exoproteome heterogeneity. Nonetheless, 30 highly conserved extracellular proteins, of which about 50% have a predicted role in pathogenesis, were dominantly identified. To approximate the virulence of the 20 investigated isolates, we employed infection models based on Galleria mellonella and HeLa cells. The results show that the grouping of clinical isolates based on their exoproteome profile can be related to virulence. We consider this outcome important as our approach provides a tool to pinpoint differences in virulence among seemingly highly similar clinical isolates of S. aureus.
Collapse
Affiliation(s)
- Xin Zhao
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Laura M Palma Medina
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Tim Stobernack
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Corinna Glasner
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Anne de Jong
- University of Groningen , Groningen Biomolecular Sciences and Biotechnology Institute, Department of Molecular Genetics , 9747 AG Groningen , The Netherlands
| | - Putri Utari
- University of Groningen , Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology , A. Deusinglaan 1 , 9713 AV Groningen , The Netherlands
| | - Rita Setroikromo
- University of Groningen , Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology , A. Deusinglaan 1 , 9713 AV Groningen , The Netherlands
| | - Wim J Quax
- University of Groningen , Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology , A. Deusinglaan 1 , 9713 AV Groningen , The Netherlands
| | - Andreas Otto
- Institut für Mikrobiologie , University of Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Dörte Becher
- Institut für Mikrobiologie , University of Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Girbe Buist
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Jan Maarten van Dijl
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| |
Collapse
|
249
|
Graf AC, Leonard A, Schäuble M, Rieckmann LM, Hoyer J, Maass S, Lalk M, Becher D, Pané-Farré J, Riedel K. Virulence Factors Produced by Staphylococcus aureus Biofilms Have a Moonlighting Function Contributing to Biofilm Integrity. Mol Cell Proteomics 2019; 18:1036-1053. [PMID: 30850421 PMCID: PMC6553939 DOI: 10.1074/mcp.ra118.001120] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/19/2019] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus is the causative agent of various biofilm-associated infections in humans causing major healthcare problems worldwide. This type of infection is inherently difficult to treat because of a reduced metabolic activity of biofilm-embedded cells and the protective nature of a surrounding extracellular matrix (ECM). However, little is known about S. aureus biofilm physiology and the proteinaceous composition of the ECM. Thus, we cultivated S. aureus biofilms in a flow system and comprehensively profiled intracellular and extracellular (ECM and flow-through (FT)) biofilm proteomes, as well as the extracellular metabolome compared with planktonic cultures. Our analyses revealed the expression of many pathogenicity factors within S. aureus biofilms as indicated by a high abundance of capsule biosynthesis proteins along with various secreted virulence factors, including hemolysins, leukotoxins, and lipases as a part of the ECM. The activity of ECM virulence factors was confirmed in a hemolysis assay and a Galleria mellonella pathogenicity model. In addition, we uncovered a so far unacknowledged moonlighting function of secreted virulence factors and ribosomal proteins trapped in the ECM: namely their contribution to biofilm integrity. Mechanistically, it was revealed that this stabilizing effect is mediated by the strong positive charge of alkaline virulence factors and ribosomal proteins in an acidic ECM environment, which is caused by the release of fermentation products like formate, lactate, and acetate because of oxygen limitation in biofilms. The strong positive charge of these proteins most likely mediates electrostatic interactions with anionic cell surface components, eDNA, and anionic metabolites. In consequence, this leads to strong cell aggregation and biofilm stabilization. Collectively, our study identified a new molecular mechanism during S. aureus biofilm formation and thus significantly widens the understanding of biofilm-associated S. aureus infections - an essential prerequisite for the development of novel antimicrobial therapies.
Collapse
Affiliation(s)
- Alexander C Graf
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Anne Leonard
- §Institute of Biochemistry, Department of Cellular Biochemistry and Metabolomics
| | - Manuel Schäuble
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Lisa M Rieckmann
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Juliane Hoyer
- ¶Institute of Microbiology, Department of Microbial Proteomics; University of Greifswald, Germany
| | - Sandra Maass
- ¶Institute of Microbiology, Department of Microbial Proteomics; University of Greifswald, Germany
| | - Michael Lalk
- §Institute of Biochemistry, Department of Cellular Biochemistry and Metabolomics
| | - Dörte Becher
- ¶Institute of Microbiology, Department of Microbial Proteomics; University of Greifswald, Germany
| | - Jan Pané-Farré
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Katharina Riedel
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology;
| |
Collapse
|
250
|
Phenotypic and Genotypic Characteristics of Methicillin-Resistant Staphylococcus aureus (MRSA) Related to Persistent Endovascular Infection. Antibiotics (Basel) 2019; 8:antibiotics8020071. [PMID: 31146412 PMCID: PMC6627527 DOI: 10.3390/antibiotics8020071] [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: 05/01/2019] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/03/2022] Open
Abstract
Persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia (PB) represents an important subset of S. aureus infection and correlates with poor clinical outcomes. MRSA isolates from patients with PB differ significantly from those of resolving bacteremia (RB) with regard to several in vitro phenotypic and genotypic profiles. For instance, PB strains exhibit less susceptibility to cationic host defense peptides and vancomycin (VAN) killing under in vivo-like conditions, greater damage to endothelial cells, thicker biofilm formation, altered growth rates, early activation of many global virulence regulons (e.g., sigB, sarA, sae and agr) and higher expression of purine biosynthesis genes (e.g., purF) than RB strains. Importantly, PB strains are significantly more resistant to VAN treatment in experimental infective endocarditis as compared to RB strains, despite similar VAN minimum inhibitory concentrations (MICs) in vitro. Here, we review relevant phenotypic and genotypic characteristics related to the PB outcome. These and future insights may improve our understanding of the specific mechanism(s) contributing to the PB outcome, and aid in the development of novel therapeutic and preventative measures against this life-threatening infection.
Collapse
|