1
|
Song L, Schwinn LS, Barthel J, Ketter V, Lechler P, Linne U, Rastan AJ, Vogt S, Ruchholtz S, Paletta JRJ, Günther M. Implant-Derived S. aureus Isolates Drive Strain-Specific Invasion Dynamics and Bioenergetic Alterations in Osteoblasts. Antibiotics (Basel) 2025; 14:119. [PMID: 40001363 PMCID: PMC11852183 DOI: 10.3390/antibiotics14020119] [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: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Implants are integral to modern orthopedic surgery. The outcomes are good, but infections remain a serious issue. Staphylococcus aureus (S. aureus), along with Staphylococcus epidermidis, are predominant pathogens responsible for implant-associated infections, as conventional antibiotic treatments often fail due to biofilm formation or the pathogens' ability to invade cells and to persist intracellularly. Objectives: This study therefore focused on interactions of S. aureus isolates from infected implants with MG63 and SaOS2 osteoblasts by investigating the adhesion, invasion, and the impact on the bioenergetics of osteoblasts. Methods and Results: We found that the ability of S. aureus to adhere to osteoblasts depends on the isolate and was not associated with a single gene or expression pattern of characteristic adhesion proteins, and further, was not correlated with invasion. However, analysis of invasion capabilities identified better invasion conditions for S. aureus isolates with the SaOS2 osteoblastic cells. Interestingly, metabolic activity of osteoblasts remained unaffected by S. aureus infection, indicating cell survival. In contrast, respiration assays revealed an altered mitochondrial bioenergetic turnover in infected cells. While basal as well as maximal respiration in MG63 osteoblasts were not influenced statistically by S. aureus infections, we found increased non-mitochondrial respiration and enhanced glycolytic activity in the osteoblasts, which was again, more pronounced in the SaOS2 osteoblastic cells. Conclusions: Our findings highlight the complexity of S. aureus-host interactions, where both the pathogen and the host cell contribute to intracellular persistence and survival, representing a major factor for therapeutic failures.
Collapse
Affiliation(s)
- Lei Song
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Lea-Sophie Schwinn
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Juliane Barthel
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Vanessa Ketter
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Philipp Lechler
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Uwe Linne
- Faculty of Chemistry, Philipps-University Marburg, 35032 Marburg, Germany
| | - Ardawan J. Rastan
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Sebastian Vogt
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Steffen Ruchholtz
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Jürgen R. J. Paletta
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Madeline Günther
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| |
Collapse
|
2
|
Kuhn EMA, Sominsky LA, Chittò M, Schwarz EM, Moriarty TF. Antibacterial Mechanisms and Clinical Impact of Sitafloxacin. Pharmaceuticals (Basel) 2024; 17:1537. [PMID: 39598446 PMCID: PMC11597390 DOI: 10.3390/ph17111537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Sitafloxacin is a 4th generation fluoroquinolone antibiotic with broad activity against a wide range of Gram-negative and Gram-positive bacteria. It is approved in Japan and used to treat pneumonia and urinary tract infections (UTIs) as well as other upper and lower respiratory infections, genitourinary infections, oral infections and otitis media. Compared to other fluoroquinolones, sitafloxacin displays a low minimal inhibitory concentration (MIC) for many bacterial species but also activity against anaerobes, intracellular bacteria, and persisters. Furthermore, it has also shown strong activity against biofilms of P. aeruginosa and S. aureus in vitro, which was recently validated in vivo with murine models of S. aureus implant-associated bone infection. Although limited in scale at present, the published literature supports the further evaluation of sitafloxacin in implant-related infections and other biofilm-related infections. The aim of this review is to summarize the chemical-positioning-based mechanisms, activity, resistance profile, and future clinical potential of sitafloxacin.
Collapse
Affiliation(s)
- Elian M. A. Kuhn
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
- Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Levy A. Sominsky
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA (E.M.S.)
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Marco Chittò
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA (E.M.S.)
| | - T. Fintan Moriarty
- AO Research Institute Davos, 7270 Davos, Switzerland; (E.M.A.K.); (M.C.)
| |
Collapse
|
3
|
Cui S, Kim E. Quorum sensing and antibiotic resistance in polymicrobial infections. Commun Integr Biol 2024; 17:2415598. [PMID: 39430726 PMCID: PMC11487952 DOI: 10.1080/19420889.2024.2415598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/20/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024] Open
Abstract
Quorum sensing (QS) is a critical bacterial communication system regulating behaviors like biofilm formation, virulence, and antibiotic resistance. This review highlights QS's role in polymicrobial infections, where bacterial species interactions enhance antibiotic resistance. We examine QS mechanisms, such as acyl-homoserine lactones (AHLs) in Gram-negative bacteria and autoinducing peptides (AIPs) in Gram-positive bacteria, and their impact on biofilm-associated antibiotic resistance. The challenges uniquely associated with polymicrobial infections, such as those found in cystic fibrosis lung infections, chronic wound infections, and medical device infections, are also summarized. Furthermore, we explore various laboratory models, including flow cells and dual-species culture models, used to study QS interactions in polymicrobial environments. The review also discusses promising quorum sensing inhibitors (QSIs), such as furanones and AHL analogs, which have demonstrated efficacy in reducing biofilm formation and virulence in laboratory and clinical studies. By addressing the interplay between QS and antibiotic resistance, this paper aims to advance therapeutic strategies that disrupt bacterial communication and improve antibiotic efficacy, ultimately mitigating the global challenge of antibiotic resistance in polymicrobial infections.
Collapse
Affiliation(s)
- Sunny Cui
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Esther Kim
- Arts and Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Burford-Gorst CM, Kidd SP. Phenotypic Variation in Staphylococcus aureus during Colonisation Involves Antibiotic-Tolerant Cell Types. Antibiotics (Basel) 2024; 13:845. [PMID: 39335018 PMCID: PMC11428495 DOI: 10.3390/antibiotics13090845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Staphylococcus aureus is a bacterial species that is commonly found colonising healthy individuals but that presents a paradoxical nature: simultaneously, it can migrate within the body and cause a range of diseases. Many of these become chronic by resisting immune responses, antimicrobial treatment, and medical intervention. In part, this ability to persist can be attributed to the adoption of multiple cell types within a single cellular population. These dynamics in the S. aureus cell population could be the result of its interplay with host cells or other co-colonising bacteria-often coagulase-negative Staphylococcal (CoNS) species. Further understanding of the unique traits of S. aureus alternative cell types, the drivers for their selection or formation during disease, as well as their presence even during non-pathological colonisation could advance the development of diagnostic tools and drugs tailored to target specific cells that are eventually responsible for chronic infections.
Collapse
Affiliation(s)
- Chloe M Burford-Gorst
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Research Centre for Infectious Diseases (RCID), The University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephen P Kidd
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Research Centre for Infectious Diseases (RCID), The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
5
|
Onyango LA, Liang J. Manuka honey as a non-antibiotic alternative against Staphylococcus spp. and their small colony variant (SCVs) phenotypes. Front Cell Infect Microbiol 2024; 14:1380289. [PMID: 38868298 PMCID: PMC11168119 DOI: 10.3389/fcimb.2024.1380289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/21/2024] [Indexed: 06/14/2024] Open
Abstract
The antibiotic resistance (ABR) crisis is an urgent global health priority. Staphylococci are among the problematic bacteria contributing to this emergency owing to their recalcitrance to many clinically important antibiotics. Staphylococcal pathogenesis is further complicated by the presence of small colony variants (SCVs), a bacterial subpopulation displaying atypical characteristics including retarded growth, prolific biofilm formation, heightened antibiotic tolerance, and enhanced intracellular persistence. These capabilities severely impede current chemotherapeutics, resulting in chronic infections, poor patient outcomes, and significant economic burden. Tackling ABR requires alternative measures beyond the conventional options that have dominated treatment regimens over the past 8 decades. Non-antibiotic therapies are gaining interest in this arena, including the use of honey, which despite having ancient therapeutic roots has now been reimagined as an alternative treatment beyond just traditional topical use, to include the treatment of an array of difficult-to-treat staphylococcal infections. This literature review focused on Manuka honey (MH) and its efficacy as an anti-staphylococcal treatment. We summarized the studies that have used this product and the technologies employed to study the antibacterial mechanisms that render MH a suitable agent for the management of problematic staphylococcal infections, including those involving staphylococcal SCVs. We also discussed the status of staphylococcal resistance development to MH and other factors that may impact its efficacy as an alternative therapy to help combat ABR.
Collapse
Affiliation(s)
- Laura A. Onyango
- Department of Biology, Trinity Western University, Langley, BC, Canada
| | | |
Collapse
|
6
|
Lo HY, Long DR, Holmes EA, Penewit K, Hodgson T, Lewis JD, Waalkes A, Salipante SJ. Transposon sequencing identifies genes impacting Staphylococcus aureus invasion in a human macrophage model. Infect Immun 2023; 91:e0022823. [PMID: 37676013 PMCID: PMC10580828 DOI: 10.1128/iai.00228-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 09/08/2023] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen in many host cell types, facilitating its persistence in chronic infections. The genes contributing to intracellular pathogenesis have not yet been fully enumerated. Here, we cataloged genes influencing S. aureus invasion and survival within human THP-1 derived macrophages using two laboratory strains (ATCC2913 and JE2). We developed an in vitro transposition method to produce highly saturated transposon mutant libraries in S. aureus and performed transposon insertion sequencing (Tn-Seq) to identify candidate genes with significantly altered abundance following macrophage invasion. While some significant genes were strain-specific, 108 were identified as common across both S. aureus strains, with most (n = 106) being required for optimal macrophage infection. We used CRISPR interference (CRISPRi) to functionally validate phenotypic contributions for a subset of genes. Of the 20 genes passing validation, seven had previously identified roles in S. aureus virulence, and 13 were newly implicated. Validated genes frequently evidenced strain-specific effects, yielding opposing phenotypes when knocked down in the alternative strain. Genomic analysis of de novo mutations occurring in groups (n = 237) of clonally related S. aureus isolates from the airways of chronically infected individuals with cystic fibrosis (CF) revealed significantly greater in vivo purifying selection in conditionally essential candidate genes than those not associated with macrophage invasion. This study implicates a core set of genes necessary to support macrophage invasion by S. aureus, highlights strain-specific differences in phenotypic effects of effector genes, and provides evidence for selection of candidate genes identified by Tn-Seq analyses during chronic airway infection in CF patients in vivo.
Collapse
Affiliation(s)
- Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elizbeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
7
|
Zhang A, Wu H, Chen X, Chen Z, Pan Y, Qu W, Hao H, Chen D, Xie S. Targeting and arginine-driven synergizing photodynamic therapy with nutritional immunotherapy nanosystems for combating MRSA biofilms. SCIENCE ADVANCES 2023; 9:eadg9116. [PMID: 37450586 PMCID: PMC10348676 DOI: 10.1126/sciadv.adg9116] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
The resistance and immune escape of methicillin-resistant Staphylococcus aureus (MRSA) biofilms cause recalcitrant infections. Here, we design a targeting and synergizing cascade PDT with nutritional immunotherapy nanosystems (Arg-PCN@Gel) containing PCN-224 as PDT platform for providing reactive oxygen species (ROS), incorporating arginine (Arg) as nitric oxide (NO) donor to cascade with ROS to produce more lethal ONOO- and promote immune response, and coating with gelatin as targeting agent and persistent Arg provider. The nanosystems adhered to the autolysin of MRSA and inhibited Arg metabolism by down-regulating icdA and icaA. It suppressed polysaccharide intercellular adhesin and extracellular DNA synthesis to prevent biofilm formation. The NO broke mature biofilms and helped ROS and ONOO- penetrate into biofilms to inactivate internal MRSA. Arg-PCN@Gel drove Arg to enhance immunity via inducible NO synthase/NO axis and arginase/polyamine axis and achieve efficient target treatment in MRSA biofilm infections. The targeting and cascading PDT synergized with nutritional immunotherapy provide an effective promising strategy for biofilm-associated infections.
Collapse
Affiliation(s)
- Aoxue Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Hao Wu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Xin Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhen Chen
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Yuanhu Pan
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Wei Qu
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Haihong Hao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Shuyu Xie
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| |
Collapse
|
8
|
Millette G, Séguin DL, Isabelle C, Chamberland S, Lucier JF, Rodrigue S, Cantin AM, Malouin F. Staphylococcus aureus Small-Colony Variants from Airways of Adult Cystic Fibrosis Patients as Precursors of Adaptive Antibiotic-Resistant Mutations. Antibiotics (Basel) 2023; 12:1069. [PMID: 37370388 PMCID: PMC10294822 DOI: 10.3390/antibiotics12061069] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Prototypic Staphylococcus aureus and their small-colony variants (SCVs) are predominant in cystic fibrosis (CF), but the interdependence of these phenotypes is poorly understood. We characterized S. aureus isolates from adult CF patients over several years. Of 18 S. aureus-positive patients (58%), 13 (72%) were positive for SCVs. Characterization included genotyping, SCCmec types, auxotrophy, biofilm production, antibiotic susceptibilities and tolerance, and resistance acquisition rates. Whole-genome sequencing revealed that several patients were colonized with prototypical and SCV-related clones. Some clonal pairs showed acquisition of aminoglycoside resistance that was not explained by aminoglycoside-modifying enzymes, suggesting a mutation-based process. The characteristics of SCVs that could play a role in resistance acquisition were thus investigated further. For instance, SCV isolates produced more biofilm (p < 0.05) and showed a higher survival rate upon exposure to ciprofloxacin and vancomycin compared to their prototypic associated clones. SCVs also developed spontaneous rifampicin resistance mutations at a higher frequency. Accordingly, a laboratory-derived SCV (ΔhemB) acquired resistance to ciprofloxacin and gentamicin faster than its parent counterpart after serial passages in the presence of sub-inhibitory concentrations of antibiotics. These results suggest a role for SCVs in the establishment of persistent antibiotic-resistant clones in adult CF patients.
Collapse
Affiliation(s)
- Guillaume Millette
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - David Lalonde Séguin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Charles Isabelle
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Suzanne Chamberland
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Jean-François Lucier
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Sébastien Rodrigue
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - André M. Cantin
- Service de Pneumologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - François Malouin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| |
Collapse
|
9
|
Sadat A, Farag AMM, Elhanafi D, Awad A, Elmahallawy EK, Alsowayeh N, El-khadragy MF, Elshopakey GE. Immunological and Oxidative Biomarkers in Bovine Serum from Healthy, Clinical, and Sub-Clinical Mastitis Caused by Escherichia coli and Staphylococcus aureus Infection. Animals (Basel) 2023; 13:ani13050892. [PMID: 36899749 PMCID: PMC10000043 DOI: 10.3390/ani13050892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
The study aimed to investigate the mastitis' emerging causative agents and their antimicrobial sensitivity, in addition to the hematological, biochemical indicators, oxidative biomarkers, acute phase protein (APP), and inflammatory cytokine changes in dairy farms in Gamasa, Dakahlia Governorate, Egypt. One hundred Holstein Friesian dairy cattle with clinical and subclinical mastitis were investigated and were allocated into three groups based on a thorough clinical examination. Escherichia coli and Staphylococcus aureus were found responsible for the clinical and subclinical mastitis in dairy farms, respectively. Multiple drug resistance (MDR) was detected in 100%, and 94.74% of E. coli and S. aureus isolates, respectively. Significantly low RBCs count, Hb, and PCV values were detected in mastitic cows compared with both subclinical mastitic and control groups; moreover, WBCs, lymphocytes, and neutrophil counts were significantly diminished in mastitic cows compared to the controls. Significantly higher levels of AST, LDH, total protein, and globulin were noticed in both mastitic and subclinical mastitic cows. The haptoglobin, fibrinogen, amyloid A, ceruloplasmin, TNF-α, IL-1β, and IL-6 levels were statistically increased in mastitic cows compared to the controls. Higher MDA levels and reduction of TAC and catalase were identified in all the mastitic cases compared to the controls. Overall, the findings suggested potential public health hazards due to antimicrobial resistance emergence. Meanwhile, the APP and cytokines, along with antioxidant markers can be used as early indicators of mastitis.
Collapse
Affiliation(s)
- Asmaa Sadat
- Department of Bacteriology, Mycology, and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence: (A.S.); (G.E.E.); Tel.: +20-1099633122 (A.S.); +20-1023923945 (G.E.E.); Fax: +20-502379952 (A.S.); +20-502379952 (G.E.E.)
| | - Alshimaa M. M. Farag
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Driss Elhanafi
- Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, NC 27606, USA
| | - Amal Awad
- Department of Bacteriology, Mycology, and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - Noorah Alsowayeh
- Department of Biology, College of Education (Majmaah), Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Manal F. El-khadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Gehad E. Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence: (A.S.); (G.E.E.); Tel.: +20-1099633122 (A.S.); +20-1023923945 (G.E.E.); Fax: +20-502379952 (A.S.); +20-502379952 (G.E.E.)
| |
Collapse
|
10
|
Functional mgrA Influences Genetic Changes within a Staphylococcus aureus Cell Population over Time. J Bacteriol 2022; 204:e0013822. [PMID: 36154359 DOI: 10.1128/jb.00138-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prolonged survival in the host-bacteria microenvironment drives the selection of alternative cell types in Staphylococcus aureus, permitting quasi-dormant sub-populations to develop. These facilitate antibiotic tolerance, long-term growth, and relapse of infection. Small Colony Variants (SCV) are an important cell type associated with persistent infection but are difficult to study in vitro due to the instability of the phenotype and reversion to the normal cell type. We have previously reported that under conditions of growth in continuous culture over a prolonged culture time, SCVs dominated a heterogenous population of cell types and these SCVs harbored a mutation in the DNA binding domain of the gene for the transcription factor, mgrA. To investigate this specific cell type further, S. aureus WCH-SK2-ΔmgrA itself was assessed with continuous culture. Compared to the wild type, the mgrA mutant strain required fewer generations to select for SCVs. There was an increased rate of mutagenesis within the ΔmgrA strain compared to the wild type, which we postulate is the mechanism explaining the increased emergence of SCV selection. The mgrA derived SCVs had impeded metabolism, altered MIC to specific antibiotics and an increased biofilm formation compared to non-SCV strain. Whole genomic sequencing detected single nucleotide polymorphisms (SNP) in phosphoglucosamine mutase glmM and tyrosine recombinase xerC. In addition, several genomic rearrangements were detected which affected genes involved in important functions such as antibiotic and toxic metal resistance and pathogenicity. Thus, we propose a direct link between mgrA and the SCV phenotype. IMPORTANCE Within a bacterial population, a stochastically generated heterogeneity of phenotypes allows continual survival against current and future stressors. The generation of a sub-population of quasi-dormant Small Colony Variants (SCV) in Staphylococcus aureus is such a mechanism, allowing for persistent or relapse of infection despite initial intervention seemingly clearing the infection. The use of continuous culture under clinically relevant conditions has allowed us to introduce time to the growth system and selects SCV within the population. This study provides valuable insights into the generation of SCV which are not addressed in standard laboratory generated models and reveals new pathways for understanding persistent S. aureus infection which can potentially be targeted in future treatments of persistent S. aureus infection.
Collapse
|
11
|
Zelmer AR, Nelson R, Richter K, Atkins GJ. Can intracellular Staphylococcus aureus in osteomyelitis be treated using current antibiotics? A systematic review and narrative synthesis. Bone Res 2022; 10:53. [PMID: 35961964 PMCID: PMC9374758 DOI: 10.1038/s41413-022-00227-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Approximately 40% of treatments of chronic and recurrent osteomyelitis fail in part due to bacterial persistence. Staphylococcus aureus, the predominant pathogen in human osteomyelitis, is known to persist by phenotypic adaptation as small-colony variants (SCVs) and by formation of intracellular reservoirs, including those in major bone cell types, reducing susceptibility to antibiotics. Intracellular infections with S. aureus are difficult to treat; however, there are no evidence-based clinical guidelines addressing these infections in osteomyelitis. We conducted a systematic review of the literature to determine the demonstrated efficacy of all antibiotics against intracellular S. aureus relevant to osteomyelitis, including protein biosynthesis inhibitors (lincosamides, streptogramins, macrolides, oxazolidines, tetracyclines, fusidic acid, and aminoglycosides), enzyme inhibitors (fluoroquinolones and ansamycines), and cell wall inhibitors (beta-lactam inhibitors, glycopeptides, fosfomycin, and lipopeptides). The PubMed and Embase databases were screened for articles related to intracellular S. aureus infections that compared the effectiveness of multiple antibiotics or a single antibiotic together with another treatment, which resulted in 34 full-text articles fitting the inclusion criteria. The combined findings of these studies were largely inconclusive, most likely due to the plethora of methodologies utilized. Therefore, the reported findings in the context of the models employed and possible solutions for improved understanding are explored here. While rifampicin, oritavancin, linezolid, moxifloxacin and oxacillin were identified as the most effective potential intracellular treatments, the scientific evidence for these is still relatively weak. We advocate for more standardized research on determining the intracellular effectiveness of antibiotics in S. aureus osteomyelitis to improve treatments and patient outcomes.
Collapse
Affiliation(s)
- Anja R Zelmer
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, SA, 5000, Australia.,Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Katharina Richter
- Richter Lab, Department of Surgery, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA, 5011, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia.
| |
Collapse
|
12
|
Causative Pathogens Do Not Differ between Early, Delayed or Late Fracture-Related Infections. Antibiotics (Basel) 2022; 11:antibiotics11070943. [PMID: 35884197 PMCID: PMC9312187 DOI: 10.3390/antibiotics11070943] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/06/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
Fracture-related infections (FRIs) are classically considered to be early (0−2 weeks), delayed (3−10 weeks) or late (>10 weeks) based on hypothesized differences in causative pathogens and biofilm formation. Treatment strategies often reflect this classification, with debridement, antimicrobial therapy and implant retention (DAIR) preferentially reserved for early FRI. This study examined pathogens isolated from FRI to confirm or refute these hypothesized differences in causative pathogens over time. Cases of FRI managed surgically at three centres between 2015−2019 and followed up for at least one year were included. Data were analysed regarding patient demographics, time from injury and pathogens isolated. Patients who underwent DAIR were also analysed separately. In total, 433 FRIs were studied, including 51 early cases (median time from injury of 2 weeks, interquartile range (IQR) of 1−2 weeks), 82 delayed cases (median time from injury of 5 weeks, IQR of 4−8 weeks) and 300 late cases (median time from injury of 112 weeks, IQR of 40−737 weeks). The type of infection was associated with time since injury; early or delayed FRI are most likely to be polymicrobial, whereas late FRIs are more likely to be culture-negative, or monomicrobial. Staphylococcus aureus was the most commonly isolated pathogen at all time points; however, we found no evidence that the type of pathogens isolated in early, delayed or late infections were different (p = 0.2). More specifically, we found no evidence for more virulent pathogens (S. aureus, Gram-negative aerobic bacilli) in early infections and less virulent pathogens (such as coagulase negative staphylococci) in late infections. In summary, decisions on FRI treatment should not assume microbiological differences related to time since injury. From a microbiological perspective, the relevance of classifying FRI by time since injury remains unclear.
Collapse
|
13
|
Bär J, Boumasmoud M, Mairpady Shambat S, Vulin C, Huemer M, Schweizer TA, Gómez-Mejia A, Eberhard N, Achermann Y, Zingg PO, Mestres C, Brugger SD, Schuepbach RA, Kouyos RD, Hasse B, Zinkernagel AS. Quantification of within-patient Staphylococcus aureus phenotypic heterogeneity as a proxy for presence of persisters across clinical presentations. Clin Microbiol Infect 2022; 28:1022.e1-1022.e7. [DOI: 10.1016/j.cmi.2022.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 12/21/2022]
|
14
|
Nurjadi D, Chanthalangsy Q, Zizmann E, Stuermer V, Moll M, Klein S, Boutin S, Heeg K, Zanger P. Phenotypic Detection of Hemin-Inducible Trimethoprim-Sulfamethoxazole Heteroresistance in Staphylococcus aureus. Microbiol Spectr 2021; 9:e0151021. [PMID: 34704796 PMCID: PMC8549745 DOI: 10.1128/spectrum.01510-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022] Open
Abstract
Trimethoprim-sulfamethoxazole (SXT) is a valuable second-line antimicrobial agent to treat methicillin-resistant Staphylococcus aureus infections. Discrepancies between various antibiotic susceptibility testing (AST) methods for SXT susceptibility in S. aureus have been described. Here, we describe a hemin-inducible heteroresistance phenotype in S. aureus. We compared the results of the Vitek 2 AST on a set of 95 S. aureus clinical isolates with broth microdilution, disk diffusion using standard Mueller-Hinton agar, and disk diffusion using Mueller-Hinton agar supplemented with 5% horse blood (MHF). To investigate the potential clinical relevance of SXT heteroresistance, an in vivo Galleria mellonella infection assay was performed. All Vitek 2 SXT-susceptible (n = 17) isolates were concordant with AST results by other methods applied in this study. In 32/78 (41%) of Vitek 2 SXT-resistant isolates, we observed a heteroresistant growth phenotype on MHF. The heteroresistance phenotype was associated with the presence of dfr genes, encoding trimethoprim resistance. The addition of a hemin-impregnated disk in a double disk diffusion method on standard Mueller-Hinton agar was able to induce growth in the SXT zone of inhibition. An in vivo infection assay with G. mellonella suggested that the SXT heteroresistance phenotype resulted in lethality similar to that of the SXT-resistant phenotype. In this study, we describe a novel hemin-inducible heteroresistance phenotype in S. aureus. This heteroresistance phenotype may be missed by standard AST methods but can be detected by performing disk diffusion using Mueller-Hinton agar supplemented with 5% horse blood, commonly used for AST of fastidious organisms. This phenomenon may partly explain the discrepancies of AST methods in determining SXT resistance in S. aureus. IMPORTANCE Staphylococcus aureus is one of most important pathogens in clinical medicine. Besides its virulence, the acquisition or emergence of resistance toward antibiotic agents, in particular to beta-lactam antibiotics (methicillin-resistant S. aureus [MRSA]), poses a major therapeutic challenge. Trimethoprim-sulfamethoxazole (SXT) is one of the effective antimicrobial agents of last resort to treat MRSA infections. Here, we report the detection of a SXT-heteroresistant phenotype which is inducible by hemin and can be detected using Mueller-Hinton agar supplemented with horse blood. Heteroresistance describes the presence or emergence of resistant subpopulations, which may potentially lead to inaccurate antibiotic susceptibility testing results and influence the success of antibiotic therapy.
Collapse
Affiliation(s)
- Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Quan Chanthalangsy
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Elfi Zizmann
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Vanessa Stuermer
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Maximilian Moll
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Sabrina Klein
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Sébastien Boutin
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Heeg
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Zanger
- Department of Infectious Diseases, Medical Microbiology, and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Global Health, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
15
|
Karami A, Farivar F, de Prinse TJ, Rabiee H, Kidd S, Sumby CJ, Bi J. Facile Multistep Synthesis of ZnO-Coated β-NaYF 4:Yb/Tm Upconversion Nanoparticles as an Antimicrobial Photodynamic Therapy for Persistent Staphylococcus aureus Small Colony Variants. ACS APPLIED BIO MATERIALS 2021; 4:6125-6136. [PMID: 35006903 DOI: 10.1021/acsabm.1c00473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Antibacterial treatment strategies using functional nanomaterials, such as photodynamic therapy, are urgently required to combat persistent Staphylococcus aureus small colony variant (SCV) bacteria. Using a stepwise approach involving thermolysis to form β-NaYF4:Yb/Tm upconversion nanoparticles (UCNPs) and surface ligand exchange with cetyltrimethylammonium bromide (CTAB), followed by zeolite imidazolate framework-8 (ZIF-8) coating and conversion to zinc oxide (ZnO), β-NaYF4:Yb/Tm@ZnO nanoparticles were synthesized. The direct synthesis of β-NaYF4:Yb/Tm@ZIF-8 UCNPs proved problematic due to the hydrophobic nature of the as-synthesized material, which was shown by zeta potential measurements using dynamic light scattering (DLS). To facilitate deposition of a ZnO coating, the zeta potentials of (i) as-synthesized UCNPs, (ii) calcined UCNPs, (iii) polyvinylpyrrolidone (PVP), and (iv) CTAB-coated UCNPs were measured, which revealed the CTAB-coated UCNPs to be the most hydrophilic and the better-dispersed form in water. β-NaYF4:Yb/Tm@ZIF-8 composites formed using the CTAB-coated UCNPs were then converted into β-NaYF4:Yb/Tm@ZnO nanoparticles by calcination under carefully controlled conditions. Photoluminescence analysis confirmed the upconversion process for the UCNP core, which allows the β-NaYF4:Yb/Tm@ZnO nanoparticles to photogenerate reactive oxygen species (ROS) when activated by near-infrared (NIR) radiation. The NIR-activated UCNPs@ZnO nanoparticles demonstrated potent efficacy against both Staphylococcus aureus (WCH-SK2) and its associated SCV form (0.67 and 0.76 log colony forming unit (CFU) reduction, respectively), which was attributed to ROS generated from the NIR activated β-NaYF4:Yb/Tm@ZnO nanoparticles.
Collapse
Affiliation(s)
- Afshin Karami
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Farzaneh Farivar
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Thomas J de Prinse
- Institute for Photonics and Advanced Sensing (IPAS), School of Physical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Hesamoddin Rabiee
- Advanced Water Management Centre, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Stephen Kidd
- Australian Centre for Antimicrobial Resistance Ecology, Research Centre for Infectious Disease, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Christopher J Sumby
- Department of Chemistry and Centre for Advanced Nanomaterials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jingxiu Bi
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
16
|
Le Masters T, Johnson S, Jeraldo PR, Greenwood-Quaintance KE, Cunningham SA, Abdel MP, Chia N, Patel R. Comparative Transcriptomic Analysis of Staphylococcus aureus Associated with Periprosthetic Joint Infection under in Vivo and in Vitro Conditions. J Mol Diagn 2021; 23:986-999. [PMID: 34098085 PMCID: PMC8351120 DOI: 10.1016/j.jmoldx.2021.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 02/25/2021] [Accepted: 05/06/2021] [Indexed: 11/15/2022] Open
Abstract
Transcriptomic analysis can provide insight as to how Staphylococcus aureus adapts to the environmental niche of periprosthetic joint infection (PJI), a challenging clinical infection. Here, in vivo RNA expression of eight S. aureus PJIs was compared with expression of the corresponding isolates in planktonic culture using a total RNA-sequencing approach. Expression varied among isolates, with a common trend showing increased expression of several ica-independent biofilm formation genes, including sdr, fnb, ebpS, and aaa; genes encoding enzymes and toxins, including coa, nuc, hlb, and hlgA/B/C; and genes facilitating acquisition of iron via the iron-binding molecule siderophore B (snb) and heme consumption protein (isd) pathways in PJI. Several antimicrobial resistance determinants were detected; although their presence correlated with phenotypic susceptibility of the associated isolates, no difference in expression between in vivo and in vitro conditions was identified.
Collapse
Affiliation(s)
- Thao Le Masters
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Stephen Johnson
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Patricio R Jeraldo
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota; Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Kerryl E Greenwood-Quaintance
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Scott A Cunningham
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Nicholas Chia
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota; Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
17
|
Bogut A, Magryś A. The road to success of coagulase-negative staphylococci: clinical significance of small colony variants and their pathogenic role in persistent infections. Eur J Clin Microbiol Infect Dis 2021; 40:2249-2270. [PMID: 34296355 PMCID: PMC8520507 DOI: 10.1007/s10096-021-04315-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/09/2021] [Indexed: 01/14/2023]
Abstract
Bacterial small colony variants represent an important aspect of bacterial variability. They are naturally occurring microbial subpopulations with distinctive phenotypic and pathogenic traits, reported for many clinically important bacteria. In clinical terms, SCVs tend to be associated with persistence in host cells and tissues and are less susceptible to antibiotics than their wild-type (WT) counterparts. The increased tendency of SCVs to reside intracellularly where they are protected against the host immune responses and antimicrobial drugs is one of the crucial aspects linking SCVs to recurrent or chronic infections, which are difficult to treat. An important aspect of the SCV ability to persist in the host is the quiescent metabolic state, reduced immune response and expression a changed pattern of virulence factors, including a reduced expression of exotoxins and an increased expression of adhesins facilitating host cell uptake. The purpose of this review is to describe in greater detail the currently available data regarding CoNS SCV and, in particular, their clinical significance and possible mechanisms by which SCVs contribute to the pathogenesis of the chronic infections. It should be emphasized that in spite of an increasing clinical significance of this group of staphylococci, the number of studies unraveling the mechanisms of CoNS SCVs formation and their impact on the course of the infectious process is still scarce, lagging behind the studies on S. aureus SCVs.
Collapse
Affiliation(s)
- Agnieszka Bogut
- Chair and Department of Medical Microbiology, Medical University of Lublin, ul. Chodźki 1, 20-093, Lublin, Poland
| | - Agnieszka Magryś
- Chair and Department of Medical Microbiology, Medical University of Lublin, ul. Chodźki 1, 20-093, Lublin, Poland.
| |
Collapse
|
18
|
Vozza EG, Mulcahy ME, McLoughlin RM. Making the Most of the Host; Targeting the Autophagy Pathway Facilitates Staphylococcus aureus Intracellular Survival in Neutrophils. Front Immunol 2021; 12:667387. [PMID: 34220813 PMCID: PMC8242348 DOI: 10.3389/fimmu.2021.667387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/28/2021] [Indexed: 12/21/2022] Open
Abstract
The success of Staphylococcus aureus as a human commensal and an opportunistic pathogen relies on its ability to adapt to several niches within the host. The innate immune response plays a key role in protecting the host against S. aureus infection; however, S. aureus adeptness at evading the innate immune system is indisputably evident. The “Trojan horse” theory has been postulated to describe a mechanism by which S. aureus takes advantage of phagocytes as a survival niche within the host to facilitate dissemination of S. aureus to secondary sites during systemic infection. Several studies have determined that S. aureus can parasitize both professional and non-professional phagocytes by manipulating the host autophagy pathway in order to create an intracellular survival niche. Neutrophils represent a critical cell type in S. aureus infection as demonstrated by the increased risk of infection among patients with congenital neutrophil disorders. However, S. aureus has been repeatedly shown to survive intracellularly within neutrophils with evidence now supporting a pathogenic role of host autophagy. By manipulating this pathway, S. aureus can also alter the apoptotic fate of the neutrophil and potentially skew other important signalling pathways for its own gain. Understanding these critical host-pathogen interactions could lead to the development of new host directed therapeutics for the treatment of S. aureus infection by removing its intracellular niche and restoring host bactericidal functions. This review discusses the current findings surrounding intracellular survival of S. aureus within neutrophils, the pathogenic role autophagy plays in this process and considers the therapeutic potential for targeting this immune evasion mechanism.
Collapse
Affiliation(s)
- Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Michelle E Mulcahy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
Greninger AL, Addetia A, Tao Y, Adler A, Qin X. Inactivation of genes in oxidative respiration and iron acquisition pathways in pediatric clinical isolates of Small colony variant Enterobacteriaceae. Sci Rep 2021; 11:7457. [PMID: 33811225 PMCID: PMC8018945 DOI: 10.1038/s41598-021-86764-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/11/2021] [Indexed: 02/01/2023] Open
Abstract
Isolation of bacterial small colony variants (SCVs) from clinical specimens is not uncommon and can fundamentally change the outcome of the associated infections. Bacterial SCVs often emerge with their normal colony phenotype (NCV) co-isolates in the same sample. The basis of SCV emergence in vivo is not well understood in Gram-negative bacteria. In this study, we interrogated the causal genetic lesions of SCV growth in three pairs of NCV and SCV co-isolates of Escherichia coli, Citrobacter freundii, and Enterobacter hormaechei. We confirmed SCV emergence was attributed to limited genomic mutations: 4 single nucleotide variants in the E. coli SCV, 5 in C. freundii, and 8 in E. hormaechei. In addition, a 10.2 kb chromosomal segment containing 11 genes was deleted in the E. hormaechei SCV isolate. Each SCV had at least one coding change in a gene associated with bacterial oxidative respiration and another involved in iron capture. Chemical and genetic rescue confirmed defects in heme biosynthesis for E. coli and C. freundii and lipoic acid biosynthesis in E. hormaachei were responsible for the SCV phenotype. Prototrophic growth in all 3 SCV Enterobacteriaceae species was unaffected under anaerobic culture conditions in vitro, illustrating how SCVs may persist in vivo.
Collapse
Affiliation(s)
- Alexander L. Greninger
- grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA ,grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| | - Amin Addetia
- grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA ,grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| | - Yue Tao
- grid.16821.3c0000 0004 0368 8293Shanghai Children’s Medical Center, Translational Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Amanda Adler
- grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| | - Xuan Qin
- grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA ,grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| |
Collapse
|
20
|
Gimza BD, Cassat JE. Mechanisms of Antibiotic Failure During Staphylococcus aureus Osteomyelitis. Front Immunol 2021; 12:638085. [PMID: 33643322 PMCID: PMC7907425 DOI: 10.3389/fimmu.2021.638085] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus is a highly successful Gram-positive pathogen capable of causing both superficial and invasive, life-threatening diseases. Of the invasive disease manifestations, osteomyelitis or infection of bone, is one of the most prevalent, with S. aureus serving as the most common etiologic agent. Treatment of osteomyelitis is arduous, and is made more difficult by the widespread emergence of antimicrobial resistant strains, the capacity of staphylococci to exhibit tolerance to antibiotics despite originating from a genetically susceptible background, and the significant bone remodeling and destruction that accompanies infection. As a result, there is a need for a better understanding of the factors that lead to antibiotic failure in invasive staphylococcal infections such as osteomyelitis. In this review article, we discuss the different non-resistance mechanisms of antibiotic failure in S. aureus. We focus on how bacterial niche and destructive tissue remodeling impact antibiotic efficacy, the significance of biofilm formation in promoting antibiotic tolerance and persister cell formation, metabolically quiescent small colony variants (SCVs), and potential antibiotic-protected reservoirs within the substructure of bone.
Collapse
Affiliation(s)
- Brittney D Gimza
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James E Cassat
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
21
|
França A, Gaio V, Lopes N, Melo LDR. Virulence Factors in Coagulase-Negative Staphylococci. Pathogens 2021; 10:170. [PMID: 33557202 PMCID: PMC7913919 DOI: 10.3390/pathogens10020170] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Coagulase-negative staphylococci (CoNS) have emerged as major pathogens in healthcare-associated facilities, being S. epidermidis, S. haemolyticus and, more recently, S. lugdunensis, the most clinically relevant species. Despite being less virulent than the well-studied pathogen S. aureus, the number of CoNS strains sequenced is constantly increasing and, with that, the number of virulence factors identified in those strains. In this regard, biofilm formation is considered the most important. Besides virulence factors, the presence of several antibiotic-resistance genes identified in CoNS is worrisome and makes treatment very challenging. In this review, we analyzed the different aspects involved in CoNS virulence and their impact on health and food.
Collapse
Affiliation(s)
- Angela França
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; (V.G.); (N.L.)
| | | | | | - Luís D. R. Melo
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; (V.G.); (N.L.)
| |
Collapse
|
22
|
Hagras M, Abutaleb NS, Elhosseiny NM, Abdelghany TM, Omara M, Elsebaei MM, Alhashimi M, Norvil AB, Gutay MI, Gowher H, Attia AS, Seleem MN, Mayhoub AS. Development of Biphenylthiazoles Exhibiting Improved Pharmacokinetics and Potent Activity Against Intracellular Staphylococcus aureus. ACS Infect Dis 2020; 6:2887-2900. [PMID: 32897045 DOI: 10.1021/acsinfecdis.0c00137] [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: 12/12/2022]
Abstract
Exploring the structure-activity relationship (SAR) at the cationic part of arylthiazole antibiotics revealed hydrazine as an active moiety. The main objective of the study is to overcome the inherited toxicity associated with the free hydrazine. A series of hydrocarbon bridges was inserted in between the groups, to separate the two amino groups. Hence, the aminomethylpiperidine-containing analog 16 was identified as a new promising antibacterial agent with efficient antibacterial and pharmacokinetic profiles. Briefly, compound 16 outperformed vancomycin in terms of the antibacterial spectrum against vancomycin-resistant staphylococcal and enterococcal strains with minimum inhibitory concentrations (MICs) ranging from 2 to 4 μg/mL, which is a faster bactericidal mode of action, completely eradicating the high staphylococcal burden within 6-8 h, and it has a unique ability to completely clear intracellular staphylococci. In addition, the initial pharmacokinetic assessment confirmed the high metabolic stability of compound 16 (biological half-life >4 h); it had a good extravascular distribution and maintained a plasma concentration higher than the average MIC value for over 12 h. Moreover, compound 16 significantly reduced MRSA burden in an in vivo MRSA skin infection mouse experiment. These attributes collectively suggest that compound 16 is a good therapeutic candidate for invasive staphylococcal and enterococcal infections. From a mechanistic point of view, compound 16 inhibited undecaprenyl diphosphate phosphatase (UppP) with an IC50 value of 29 μM.
Collapse
Affiliation(s)
- Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Nader S. Abutaleb
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
| | - Noha M. Elhosseiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Tamer M. Abdelghany
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Mariam Omara
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Mohamed M. Elsebaei
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
| | - Allison B Norvil
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mark I Gutay
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana 47907, United States
| | - Humaira Gowher
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ahmed S. Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, United States
| | - Abdelrahman S. Mayhoub
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
- University of Science and Technology, Nanoscience Program, Zewail City of Science and Technology, Ahmed Zewail Street, October Gardens, sixth of October, Giza 12578, Egypt
| |
Collapse
|
23
|
Assoni L, Milani B, Carvalho MR, Nepomuceno LN, Waz NT, Guerra MES, Converso TR, Darrieux M. Resistance Mechanisms to Antimicrobial Peptides in Gram-Positive Bacteria. Front Microbiol 2020; 11:593215. [PMID: 33193264 PMCID: PMC7609970 DOI: 10.3389/fmicb.2020.593215] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023] Open
Abstract
With the alarming increase of infections caused by pathogenic multidrug-resistant bacteria over the last decades, antimicrobial peptides (AMPs) have been investigated as a potential treatment for those infections, directly through their lytic effect or indirectly, due to their ability to modulate the immune system. There are still concerns regarding the use of such molecules in the treatment of infections, such as cell toxicity and host factors that lead to peptide inhibition. To overcome these limitations, different approaches like peptide modification to reduce toxicity and peptide combinations to improve therapeutic efficacy are being tested. Human defense peptides consist of an important part of the innate immune system, against a myriad of potential aggressors, which have in turn developed different ways to overcome the AMPs microbicidal activities. Since the antimicrobial activity of AMPs vary between Gram-positive and Gram-negative species, so do the bacterial resistance arsenal. This review discusses the mechanisms exploited by Gram-positive bacteria to circumvent killing by antimicrobial peptides. Specifically, the most clinically relevant genera, Streptococcus spp., Staphylococcus spp., Enterococcus spp. and Gram-positive bacilli, have been explored.
Collapse
Affiliation(s)
- Lucas Assoni
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Barbara Milani
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Marianna Ribeiro Carvalho
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lucas Natanael Nepomuceno
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Natalha Tedeschi Waz
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Maria Eduarda Souza Guerra
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Thiago Rojas Converso
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Michelle Darrieux
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| |
Collapse
|
24
|
Bär J, Boumasmoud M, Kouyos RD, Zinkernagel AS, Vulin C. Efficient microbial colony growth dynamics quantification with ColTapp, an automated image analysis application. Sci Rep 2020; 10:16084. [PMID: 32999342 PMCID: PMC7528005 DOI: 10.1038/s41598-020-72979-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Populations of genetically identical bacteria are phenotypically heterogeneous, giving rise to population functionalities that would not be possible in homogeneous populations. For instance, a proportion of non-dividing bacteria could persist through antibiotic challenges and secure population survival. This heterogeneity can be studied in complex environmental or clinical samples by spreading the bacteria on agar plates and monitoring time to growth resumption in order to infer their metabolic state distribution. We present ColTapp, the Colony Time-lapse application for bacterial colony growth quantification. Its intuitive graphical user interface allows users to analyze time-lapse images of agar plates to monitor size, color and morphology of colonies. Additionally, images at isolated timepoints can be used to estimate lag time. Using ColTapp, we analyze a dataset of Staphylococcus aureus time-lapse images including populations with heterogeneous lag time. Colonies on dense plates reach saturation early, leading to overestimation of lag time from isolated images. We show that this bias can be corrected by taking into account the area available to each colony on the plate. We envision that in clinical settings, improved analysis of colony growth dynamics may help treatment decisions oriented towards personalized antibiotic therapies.
Collapse
Affiliation(s)
- Julian Bär
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mathilde Boumasmoud
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Roger D Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Clément Vulin
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland. .,Institute of Biogeochemistry and Pollutant Dynamics, ETH Zurich, 8092, Zurich, Switzerland. .,Department of Environmental Microbiology, 8600, Eawag, Dubendorf, Switzerland.
| |
Collapse
|
25
|
Miller LS, Fowler VG, Shukla SK, Rose WE, Proctor RA. Development of a vaccine against Staphylococcus aureus invasive infections: Evidence based on human immunity, genetics and bacterial evasion mechanisms. FEMS Microbiol Rev 2020; 44:123-153. [PMID: 31841134 PMCID: PMC7053580 DOI: 10.1093/femsre/fuz030] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Invasive Staphylococcus aureus infections are a leading cause of morbidity and mortality in both hospital and community settings, especially with the widespread emergence of virulent and multi-drug resistant methicillin-resistant S. aureus strains. There is an urgent and unmet clinical need for non-antibiotic immune-based approaches to treat these infections as the increasing antibiotic resistance is creating a serious threat to public health. However, all vaccination attempts aimed at preventing S. aureus invasive infections have failed in human trials, especially all vaccines aimed at generating high titers of opsonic antibodies against S. aureus surface antigens to facilitate antibody-mediated bacterial clearance. In this review, we summarize the data from humans regarding the immune responses that protect against invasive S. aureus infections as well as host genetic factors and bacterial evasion mechanisms, which are important to consider for the future development of effective and successful vaccines and immunotherapies against invasive S. aureus infections in humans. The evidence presented form the basis for a hypothesis that staphylococcal toxins (including superantigens and pore-forming toxins) are important virulence factors, and targeting the neutralization of these toxins are more likely to provide a therapeutic benefit in contrast to prior vaccine attempts to generate antibodies to facilitate opsonophagocytosis.
Collapse
Affiliation(s)
- Lloyd S Miller
- Immunology, Janssen Research and Development, 1400 McKean Road, Spring House, PA, 19477, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Cancer Research Building 2, Suite 209, Baltimore, MD, 21231, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, 1830 East Monument Street, Baltimore, MD, 21287, USA.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, 601 North Caroline Street, Baltimore, MD, 21287, USA.,Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Vance G Fowler
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, 315 Trent Drive, Hanes House, Durham, NC, 27710, USA.,Duke Clinical Research Institute, Duke University Medical Center, 40 Duke Medicine Circle, Durham, NC, 27710, USA
| | - Sanjay K Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, 1000 North Oak Avenue, Marshfield, WI, 54449, USA.,Computation and Informatics in Biology and Medicine, University of Wisconsin, 425 Henry Mall, Room 3445, Madison, WI, 53706, USA
| | - Warren E Rose
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA.,Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 4123 Rennebohm Hall, Madison, WI, 53705 USA
| | - Richard A Proctor
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA.,Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, 1550 Linden Drive, Microbial Sciences Building, Room 1334, Madison, WI, 53705, USA
| |
Collapse
|
26
|
Sulabh S, Panigrahi M, Varshney R, Gupta JP, Kumar S, Verma A, Kumar A, Asaf VM, Kumar P, Bhushan B. In-vitroanalysis of Interleukin-10 expression in cell cultures of Crossbred cattle, Tharparkar cattle and Murrah buffalo in response to mastitis causing antigens derived fromStaphylococcus aureusandEscherichia coli. BIOL RHYTHM RES 2019. [DOI: 10.1080/09291016.2019.1628407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sourabh Sulabh
- Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - Manjit Panigrahi
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - Rajat Varshney
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Bareilly, UP, India
| | - Jay P. Gupta
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - Satish Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
- Department of Animal Genetics and Breeding, ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, India
| | - Ankita Verma
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - Amod Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - V.N. Muhasin Asaf
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - Pushpendra Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| | - Bharat Bhushan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Bareilly, UP, India
| |
Collapse
|
27
|
ClpC affects the intracellular survival capacity of Staphylococcus aureus in non-professional phagocytic cells. Sci Rep 2019; 9:16267. [PMID: 31700127 PMCID: PMC6838064 DOI: 10.1038/s41598-019-52731-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
Invasion and persistence of bacteria within host cells requires that they adapt to life in an intracellular environment. This adaptation induces bacterial stress through events such as phagocytosis and enhanced nutrient-restriction. During stress, bacteria synthesize a family of proteins known as heat shock proteins (HSPs) to facilitate adaptation and survival. Previously, we determined the Staphylococcus aureus HSP ClpC temporally alters bacterial metabolism and persistence. This led us to hypothesize that ClpC might alter intracellular survival. Inactivation of clpC in S. aureus strain DSM20231 significantly enhanced long-term intracellular survival in human epithelial (HaCaT) and endothelial (EA.hy926) cell lines, without markedly affecting adhesion or invasion. This phenotype was similar across a genetically diverse collection of S. aureus isolates, and was influenced by the toxin/antitoxin encoding locus mazEF. Importantly, MazEF alters mRNA synthesis and/or stability of S. aureus virulence determinants, indicating ClpC may act through the mRNA modulatory activity of MazEF. Transcriptional analyses of total RNAs isolated from intracellular DSM20231 and isogenic clpC mutant cells identified alterations in transcription of α-toxin (hla), protein A (spa), and RNAIII, consistent with the hypothesis that ClpC negatively affects the intracellular survival of S. aureus in non-professional phagocytic cells, via modulation of MazEF and Agr.
Collapse
|
28
|
Soares A, Caron F, Etienne M. Commentary: Tolerance and Resistance of Pseudomonas aeruginosa Biofilms to Antimicrobial Agents-How P. aeruginosa Can Escape Antibiotics. Front Microbiol 2019; 10:2164. [PMID: 31620114 PMCID: PMC6759825 DOI: 10.3389/fmicb.2019.02164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/03/2019] [Indexed: 01/13/2023] Open
Affiliation(s)
- Anaïs Soares
- GRAM 2.0, EA 2656, Normandie Univ, UNIROUEN, Rouen, France.,Microbiology Department, Rouen University Hospital, Rouen, France
| | - François Caron
- GRAM 2.0, EA 2656, Normandie Univ, UNIROUEN, Rouen, France.,Infectious Diseases Department, Rouen University Hospital, Rouen, France
| | - Manuel Etienne
- GRAM 2.0, EA 2656, Normandie Univ, UNIROUEN, Rouen, France.,Infectious Diseases Department, Rouen University Hospital, Rouen, France
| |
Collapse
|
29
|
Unstable chromosome rearrangements in Staphylococcus aureus cause phenotype switching associated with persistent infections. Proc Natl Acad Sci U S A 2019; 116:20135-20140. [PMID: 31527262 PMCID: PMC6778178 DOI: 10.1073/pnas.1904861116] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen known to exhibit subpopulations of small-colony variants (SCVs) that cause persistent or recurrent infections. The underlying mechanisms promoting the SCV phenotypic switching and adaptation to persistent infection are poorly understood. Moreover, the instability of this frequently reverting phenotype hampers diagnosis and study. Here we show that SCVs with reduced virulence but increased immune evasion and persistence properties can arise from reversible chromosomal instability. This mechanism of SCV generation implies an asymmetric chromosome inversion and the activation of prophage-encoding genes used for immune evasion. Assessment of major S. aureus lineages indicates this genomic plasticity is a common but previously unrecognized mechanism used by S. aureus to cause persistent and relapsing infections. Staphylococcus aureus small-colony variants (SCVs) are associated with unusually chronic and persistent infections despite active antibiotic treatment. The molecular basis for this clinically important phenomenon is poorly understood, hampered by the instability of the SCV phenotype. Here we investigated the genetic basis for an unstable S. aureus SCV that arose spontaneously while studying rifampicin resistance. This SCV showed no nucleotide differences across its genome compared with a normal-colony variant (NCV) revertant, yet the SCV presented the hallmarks of S. aureus linked to persistent infection: down-regulation of virulence genes and reduced hemolysis and neutrophil chemotaxis, while exhibiting increased survival in blood and ability to invade host cells. Further genome analysis revealed chromosome structural variation uniquely associated with the SCV. These variations included an asymmetric inversion across half of the S. aureus chromosome via recombination between type I restriction modification system (T1RMS) genes, and the activation of a conserved prophage harboring the immune evasion cluster (IEC). Phenotypic reversion to the wild-type–like NCV state correlated with reversal of the chromosomal inversion (CI) and with prophage stabilization. Further analysis of 29 complete S. aureus genomes showed strong signatures of recombination between hsdMS genes, suggesting that analogous CI has repeatedly occurred during S. aureus evolution. Using qPCR and long-read amplicon deep sequencing, we detected subpopulations with T1RMS rearrangements causing CIs and prophage activation across major S. aureus lineages. Here, we have discovered a previously unrecognized and widespread mechanism of reversible genomic instability in S. aureus associated with SCV generation and persistent infections.
Collapse
|
30
|
Selective pressures during chronic infection drive microbial competition and cooperation. NPJ Biofilms Microbiomes 2019; 5:16. [PMID: 31263568 PMCID: PMC6555799 DOI: 10.1038/s41522-019-0089-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic infections often contain complex mixtures of pathogenic and commensal microorganisms ranging from aerobic and anaerobic bacteria to fungi and viruses. The microbial communities present in infected tissues are not passively co-existing but rather actively interacting with each other via a spectrum of competitive and/or cooperative mechanisms. Competition versus cooperation in these microbial interactions can be driven by both the composition of the microbial community as well as the presence of host defense strategies. These interactions are typically mediated via the production of secreted molecules. In this review, we will explore the possibility that microorganisms competing for nutrients at the host–pathogen interface can evolve seemingly cooperative mechanisms by controlling the production of subsets of secreted virulence factors. We will also address interspecies versus intraspecies utilization of community resources and discuss the impact that this phenomenon might have on co-evolution at the host–pathogen interface.
Collapse
|
31
|
Alreshidi MM, Dunstan RH, Macdonald MM, Smith ND, Gottfries J, Roberts TK. Amino acids and proteomic acclimation of Staphylococcus aureus when incubated in a defined minimal medium supplemented with 5% sodium chloride. Microbiologyopen 2019; 8:e00772. [PMID: 30739392 PMCID: PMC6562129 DOI: 10.1002/mbo3.772] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 11/06/2022] Open
Abstract
Staphylococcus aureus is a versatile bacterium that can adapt to survive and grow in a wide range of salt concentrations. This study investigated whether the cells could mount a response to survive a challenge of 5% NaCl in a minimal incubation medium that would not support cell replication. Cells were grown in liquid culture, washed and then incubated for 90 min at 37°C in a medium that contained only glycine and glucose as substrates in PBS plus trace elements. The control cells were compared with a treatment group which was incubated with an additional 5% NaCl. Significantly more glycine was taken up by the cells exposed to 5% NaCl compared with control cells, and both groups consumed 99% of the glucose supplied. The NaCl treated cells had significantly higher cytoplasmic levels of proline and glutamic acid as well as lower levels of alanine and methionine compared with the controls (p < 0.05). The levels of the two major cytoplasmic amino acids, aspartic acid and glycine, remained constant in control and treated cells. Proteomic analyses revealed that 10 proteins showed differential responses between the control and treatment groups. The reductions in proteins were primarily associated with processes of protein biosynthesis, pathogenicity, and cell adhesion. Since cell numbers remained constant during the incubation period in minimal medium, it was concluded that there was no cell division to support population growth. The results provided evidence that the cells in the minimal medium exposed to the NaCl treatment underwent in situ homeostatic changes to adjust to the new environmental conditions. It was proposed that this represented a phenotypic shift to form cells akin to small colony variants, with lower metabolic rates and lower levels of key proteins associated with pathogenicity.
Collapse
Affiliation(s)
- Mousa M. Alreshidi
- Department of Biology, College of ScienceUniversity of HailHailSaudi Arabia
| | - R. Hugh Dunstan
- Metabolic Research Group, Faculty of ScienceSchool of Environmental and Life SciencesCallaghanNew South WalesAustralia
| | - Margaret M. Macdonald
- Metabolic Research Group, Faculty of ScienceSchool of Environmental and Life SciencesCallaghanNew South WalesAustralia
| | - Nathan D. Smith
- Analytical and Biomolecular Research Facility (ABRF)University of NewcastleCallaghanNew South WalesAustralia
| | - Johan Gottfries
- Department of ChemistryGothenburg UniversityGothenburgSweden
| | - Tim K. Roberts
- Metabolic Research Group, Faculty of ScienceSchool of Environmental and Life SciencesCallaghanNew South WalesAustralia
| |
Collapse
|
32
|
Perez K, Patel R. Staphylococcusepidermidis Small-Colony Variants Are Induced by Low pH and Their Frequency Reduced by Lysosomal Alkalinization. J Infect Dis 2019; 215:488-490. [PMID: 27974434 DOI: 10.1093/infdis/jiw503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/13/2016] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, and.,Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Zhang P, Wright JA, Tymon A, Nair SP. Bicarbonate induces high-level resistance to the human antimicrobial peptide LL-37 in Staphylococcus aureus small colony variants. J Antimicrob Chemother 2019; 73:615-619. [PMID: 29211886 PMCID: PMC5890704 DOI: 10.1093/jac/dkx433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/25/2017] [Indexed: 01/29/2023] Open
Abstract
Objectives Staphylococcus aureus small colony variants (SCVs) cause persistent infections and are resistant to cationic antibiotics. Antimicrobial peptides (AMPs) have been suggested as promising alternatives for treating antibiotic-resistant bacteria. We investigated the capacity of the human cationic AMP LL-37 to kill SCVs in the presence of physiological concentrations of bicarbonate, which are reported to alter bacterial membrane permeability and change resistance of bacteria to AMPs. Methods MBCs of LL-37 for S. aureus SCVs with mutations in different genes in the presence and absence of bicarbonate were determined. Results In the absence of bicarbonate, SCVs of S. aureus strains LS-1 and 8325-4 had the same level of resistance to LL-37 as the parental strain (8 mg/L). In the presence of bicarbonate, hemB, menD and aroD SCVs of LS-1 had high-level resistance to LL-37 (≥128 mg/L) compared with the parental strain (16 mg/L). However, only the aroD SCV of strain 8324-5 showed high-level resistance. 8325-4 harbours mutations in two genes, tcaR and rsbU, which are involved in antimicrobial sensing and the stress response, respectively. When rsbU was repaired in 8325-4 it displayed high-level resistance to LL-37 in the presence of bicarbonate. This phenotype was lost when tcaR was also repaired, demonstrating that RsbU and TcaR are involved in LL-37 resistance in the presence of bicarbonate Conclusions S. aureus SCVs would be resistant to high concentrations of LL-37 in niches where there are physiological concentrations of bicarbonate and therefore this AMP may not be effective in combating SCVs.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Microbial Diseases, UCL Eastman Dental Institute, 256 Gray's Inn Road, London WC1X 8LD, UK
| | - John A Wright
- Immunology Catalyst, GSK, Stevenage, Hertfordshire, UK
| | - Anna Tymon
- Department of Microbial Diseases, UCL Eastman Dental Institute, 256 Gray's Inn Road, London WC1X 8LD, UK
| | - Sean P Nair
- Department of Microbial Diseases, UCL Eastman Dental Institute, 256 Gray's Inn Road, London WC1X 8LD, UK
| |
Collapse
|
34
|
General Assembly, Prevention, Local Antimicrobials: Proceedings of International Consensus on Orthopedic Infections. J Arthroplasty 2019; 34:S75-S84. [PMID: 30352772 DOI: 10.1016/j.arth.2018.09.056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
35
|
Perez K, Patel R. Survival of Staphylococcus epidermidis in Fibroblasts and Osteoblasts. Infect Immun 2018; 86:e00237-18. [PMID: 30061380 PMCID: PMC6204734 DOI: 10.1128/iai.00237-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022] Open
Abstract
Staphylococcus epidermidis is a leading cause of infections associated with indwelling medical devices, including prosthetic joint infection. While biofilm formation is assumed to be the main mechanism underlying the chronic infections S. epidermidis causes, we hypothesized that S. epidermidis also evades immune killing, contributing to its pathogenesis. Here, we show that prosthetic joint-associated S. epidermidis isolates can persist intracellularly within human fibroblasts and inside human and mouse osteoblasts. We also show that the intracellularly persisting bacteria reside primarily within acidic phagolysosomes and that over the course of infection, small-colony variants are selected for. Moreover, upon eukaryotic cell death, these bacteria, which can outlive their host, can escape into the extracellular environment, providing them an opportunity to form biofilms on implant surfaces at delayed time points in implant-associated infection. In summary, the acidic phagolysosomes of fibroblasts and osteoblasts serve as reservoirs for chronic or delayed S. epidermidis infection.
Collapse
Affiliation(s)
- Kimberly Perez
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
36
|
Sun A, Zhang H, Pang F, Niu G, Chen J, Chen F, Zhang J. Essential genes of the macrophage response to Staphylococcus aureus exposure. Cell Mol Biol Lett 2018; 23:25. [PMID: 29849669 PMCID: PMC5966896 DOI: 10.1186/s11658-018-0090-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/06/2018] [Indexed: 12/15/2022] Open
Abstract
Background Although significant advances have been made in understanding the mechanisms of macrophage response to Staphylococcus aureus infection, the molecular details are still elusive. Identification of the essential genes and biological processes of macrophages that are specifically changed at different durations of S. aureus exposure is of great clinical significance. Methods We aimed to identify the significantly changed genes and biological processes of S. aureus-exposed macrophages. We systematically analyzed the macrophage gene expression profile GSE 13670 database with 8 h, 24 h or 48 h S. aureus infection. The results were further confirmed by western blot and quantitative polymerase chain reaction (qPCR) analyses. Results After 8 h of S. aureus infection, the expression of 624 genes was significantly changed. Six hundred thirteen differentially expressed genes (DEGs) were identified after 24 h of S. aureus infection. Two hundred fifty-three genes were significantly changed after 48 h of S. aureus infection. STAT1 was consistently up-regulated in these three treatments. TP53, JAK2, CEBPA, STAT3, MYC, CTNNB1 and PRKCA were only identified in the 8 h or 24 h S. aureus infection groups. CTNNB1 and PRKCA were for the first time identified as potential essential genes in S. aureus infection of macrophages. In the Gene Ontology (GO) term analysis, the defense response was shown to be the most significantly changed biological process among all processes; KEGG pathway analysis identified the JAK-STAT signaling pathway involved in early infection. Conclusions Our systematic analysis identified unique gene expression profiles and specifically changed biological processes of the macrophage response to different S. aureus exposure times.
Collapse
Affiliation(s)
- Aixia Sun
- 1Department of Clinical Laboratory, Liaocheng People's Hospital, 67 West Dongchang Road, Liaocheng, 252000 Shandong Province People's Republic of China
| | - Hongwei Zhang
- 1Department of Clinical Laboratory, Liaocheng People's Hospital, 67 West Dongchang Road, Liaocheng, 252000 Shandong Province People's Republic of China
| | - Feng Pang
- 1Department of Clinical Laboratory, Liaocheng People's Hospital, 67 West Dongchang Road, Liaocheng, 252000 Shandong Province People's Republic of China
| | - Guifen Niu
- 2Department of Endocrinology, Liaocheng People's Hospital, 67 West Dongchang Road, Liaocheng, 252000 Shandong Province People's Republic of China
| | - Jianzhong Chen
- 3Department of Clinical Pharmacy, Liaocheng People's Hospital, 67 West Dongchang Road, Liaocheng, 252000 Shandong Province People's Republic of China
| | - Fei Chen
- 3Department of Clinical Pharmacy, Liaocheng People's Hospital, 67 West Dongchang Road, Liaocheng, 252000 Shandong Province People's Republic of China
| | - Jian Zhang
- Outpatient Vaccination Service, Center for Disease Control and Prevention of Liaocheng, Liaocheng, 252000 Shandong Province People's Republic of China
| |
Collapse
|
37
|
Murphy GR, Dunstan RH, Macdonald MM, Gottfries J, Roberts TK. Alterations in amino acid metabolism during growth by Staphylococcus aureus following exposure to H 2O 2 - A multifactorial approach. Heliyon 2018; 4:e00620. [PMID: 29756075 PMCID: PMC5944418 DOI: 10.1016/j.heliyon.2018.e00620] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/20/2018] [Accepted: 04/27/2018] [Indexed: 01/02/2023] Open
Abstract
Temperature and pH are known to vary in a wound site due to the immune response and subsequent healing processes. This study used a multifactorial design to examine the cellular responses of Staphylococcus aureus to hydrogen peroxide (0–100 mM) when bacteria were grown in temperatures of 37 ± 2 °C and pH 7 ± 1, conditions potentially encountered in wound sites. A centroid sample was included in the design which represented the mid-point values of all three environmental parameters (37 °C, pH 7, 50 mM H2O2). Cytoplasmic extracts and corresponding medium supernatants were analysed for amino acid composition by gas chromatography. Exposures of S. aureus to H2O2 during the inoculation process resulted in extended lag phases lasting well after the peroxide had been neutralised by the bacterium's antioxidant systems, after which the bacteria eventually resumed growth at equivalent rates to the controls. Even though the subsequent growth rates appeared normal, the cells exhibited a variant metabolic regime at the mid-exponential phase of growth as a result of the initial exposure to peroxide. The alterations in metabolism were reflected by the differential amino acid profiles measured in the cytoplasmic extracts (P < 0.0001). The data indicated that the metabolic responses to H2O2 challenge were uniquely different depending on the variations of temperature and pH. The uptake patterns of amino acids from the media also altered depending on prevailing environmental conditions. From these results, it was proposed that a specific reproducible homeostasis could be induced under a specific set of defined environmental conditions. It was also evident that early toxic insults on the bacterial culture could have lasting impacts on cellular homeostasis after successive generations, even after the offending chemical had been removed and initial cell integrity restored. It was concluded that metabolic homeostasis would be continually adjusting and responding to changing environmental conditions to deploy defensive proteins as well as optimising processes for survival. The powerful ability to continually and rapidly adapt to the environment may represent the key feature supporting the virulence of S. aureus as an opportunistic pathogen invading the wound site.
Collapse
Affiliation(s)
- Grace R Murphy
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| | - R Hugh Dunstan
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| | - Margaret M Macdonald
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| | | | - Tim K Roberts
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| |
Collapse
|
38
|
Belfield K, Bayston R, Hajduk N, Levell G, Birchall JP, Daniel M. Evaluation of combinations of putative anti-biofilm agents and antibiotics to eradicate biofilms of Staphylococcus aureus and Pseudomonas aeruginosa. J Antimicrob Chemother 2018; 72:2531-2538. [PMID: 28859444 DOI: 10.1093/jac/dkx192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/23/2017] [Indexed: 12/31/2022] Open
Abstract
Objectives To evaluate potential anti-biofilm agents for their ability to enhance the activity of antibiotics for local treatment of localized biofilm infections. Methods Staphylococcus aureus and Pseudomonas aeruginosa in vitro biofilm models were developed. The putative antibiotic enhancers N-acetylcysteine, acetylsalicylic acid, sodium salicylate, recombinant human deoxyribonuclease I, dispersin B, hydrogen peroxide and Johnson's Baby Shampoo (JBS) were tested for their anti-biofilm activity alone and their ability to enhance the activity of antibiotics for 7 or 14 days, against 5 day old biofilms. The antibiotic enhancers were paired with rifampicin and clindamycin against S. aureus and gentamicin and ciprofloxacin against P. aeruginosa. Isolates from biofilms that were not eradicated were tested for antibiotic resistance. Results Antibiotic levels 10× MIC and 100× MIC significantly reduced biofilm, but did not consistently eradicate it. Antibiotics at 100× MIC with 10% JBS for 14 days was the only treatment to eradicate both staphylococcal and pseudomonal biofilms. Recombinant human deoxyribonuclease I significantly reduced staphylococcal biofilm. Emergence of resistance of surviving isolates was minimal and was often associated with the small colony variant phenotype. Conclusions JBS enhanced the activity of antibiotics and several other promising anti-biofilm agents were identified. Antibiotics with 10% JBS eradicated biofilms produced by both organisms. Such combinations might be useful in local treatment of localized biofilm infections.
Collapse
Affiliation(s)
- Katherine Belfield
- Biomaterials-Related Infection Group, Department of Orthopaedics, Trauma and Sports Medicine, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Roger Bayston
- Biomaterials-Related Infection Group, Department of Orthopaedics, Trauma and Sports Medicine, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Nadzieja Hajduk
- Biomaterials-Related Infection Group, Department of Orthopaedics, Trauma and Sports Medicine, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Georgia Levell
- Biomaterials-Related Infection Group, Department of Orthopaedics, Trauma and Sports Medicine, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - John P Birchall
- NIHR Nottingham Hearing Biomedical Research Unit, Nottingham University Hospitals, Ropewalk House, Nottingham, UK.,Otology and Hearing Group, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK.,Otorhinolaryngology, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Matija Daniel
- NIHR Nottingham Hearing Biomedical Research Unit, Nottingham University Hospitals, Ropewalk House, Nottingham, UK.,Otology and Hearing Group, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK.,Otorhinolaryngology, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
39
|
Groll JÜRGEN, Fiedler JÖRG, Bruellhoff K, Moeller M, Brenner RE. Novel Surface Coatings Modulating Eukaryotic Cell Adhesion and Preventing Implant Infection. Int J Artif Organs 2018; 32:655-62. [DOI: 10.1177/039139880903200915] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Poor osseointegration and bacterial infection are major causes of orthopedic implant failure. Both problems arise from passive unspecific protein coating that may not optimally support adhesion of osteoblastic cells and which enable bacterial adhesion that subsequently results in biofilm formation. This review addresses emerging concepts of preventing unspecific protein adsorption and biofilm formation by organic coating systems. We especially focus on recent concepts that additionally allow functionalization for preferential cell adhesion using cell adhesion mediating small peptide sequences that do not induce bacterial adherence. One promising approach that is presented and discussed within this context is the use of NCO-sP(EO-stat-PO).
Collapse
Affiliation(s)
- JÜRGEN Groll
- DWI e.V. and Institute of Technical and Macromolecular Chemistry, RWTH Aachen, Aachen - Germany
| | - JÖRG Fiedler
- Department of Orthopedics, Division for Biochemistry of Joint and Connective Tissue Diseases, University of Ulm, Ulm - Germany
| | - Kristina Bruellhoff
- DWI e.V. and Institute of Technical and Macromolecular Chemistry, RWTH Aachen, Aachen - Germany
| | - Martin Moeller
- DWI e.V. and Institute of Technical and Macromolecular Chemistry, RWTH Aachen, Aachen - Germany
| | - Rolf E. Brenner
- Department of Orthopedics, Division for Biochemistry of Joint and Connective Tissue Diseases, University of Ulm, Ulm - Germany
| |
Collapse
|
40
|
Influence of IS 256 on Genome Variability and Formation of Small-Colony Variants in Staphylococcus aureus. Antimicrob Agents Chemother 2017; 61:AAC.00144-17. [PMID: 28584147 DOI: 10.1128/aac.00144-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus has acquired resistance to nearly all antibiotics used in clinical practice. Whereas some resistance mechanisms are conferred by uptake of resistance genes, others evolve by mutation. In this study, IS256 has been shown to play a role, e.g., in S. aureus strains displaying intermediate resistance to vancomycin (VISA). To characterize the IS256 insertion sites in the genomes of two closely related sequence type 247 (ST247) VISA strains, all insertions were mapped in both VISA and a susceptible control strain. The results showed that the three ST247 strains contained the highest number so far of IS256 insertions for all sequenced S. aureus strains. Furthermore, in contrast to the case with the other IS elements in these genomes, the IS256 insertion sites were not identical in the closely related strains, indicating a high transposition frequency of IS256 When IS256 was introduced into a laboratory strain which was then cultured in the presence of antibiotics, it was possible to isolate small-colony variants (SCVs) that possessed IS256 insertions in guaA and hemY that displayed increased resistance to vancomycin and aminoglycosides, respectively. For these clones, a very rapid reversion to the wild type that resembled the fast reversion of clinical SCVs was observed. The reversion was caused by excision of IS256 in a small number of fast-growing clones that quickly outcompeted the SCVs in broth cultures. In conclusion, the presence of IS256 confers a strong genomic plasticity that is useful for adaptation to antibiotic stress.
Collapse
|
41
|
Dumville JC, Lipsky BA, Hoey C, Cruciani M, Fiscon M, Xia J, Cochrane Wounds Group. Topical antimicrobial agents for treating foot ulcers in people with diabetes. Cochrane Database Syst Rev 2017; 6:CD011038. [PMID: 28613416 PMCID: PMC6481886 DOI: 10.1002/14651858.cd011038.pub2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND People with diabetes are at high risk for developing foot ulcers, which often become infected. These wounds, especially when infected, cause substantial morbidity. Wound treatments should aim to alleviate symptoms, promote healing, and avoid adverse outcomes, especially lower extremity amputation. Topical antimicrobial therapy has been used on diabetic foot ulcers, either as a treatment for clinically infected wounds, or to prevent infection in clinically uninfected wounds. OBJECTIVES To evaluate the effects of treatment with topical antimicrobial agents on: the resolution of signs and symptoms of infection; the healing of infected diabetic foot ulcers; and preventing infection and improving healing in clinically uninfected diabetic foot ulcers. SEARCH METHODS We searched the Cochrane Wounds Specialised Register, CENTRAL, Ovid MEDLINE, Ovid MEDLINE (In-Process & Other Non-Indexed Citations), Ovid Embase, and EBSCO CINAHL Plus in August 2016. We also searched clinical trials registries for ongoing and unpublished studies, and checked reference lists to identify additional studies. We used no restrictions with respect to language, date of publication, or study setting. SELECTION CRITERIA We included randomised controlled trials conducted in any setting (inpatient or outpatient) that evaluated topical treatment with any type of solid or liquid (e.g., cream, gel, ointment) antimicrobial agent, including antiseptics, antibiotics, and antimicrobial dressings, in people with diabetes mellitus who were diagnosed with an ulcer or open wound of the foot, whether clinically infected or uninfected. DATA COLLECTION AND ANALYSIS Two review authors independently performed study selection, 'Risk of bias' assessment, and data extraction. Initial disagreements were resolved by discussion, or by including a third review author when necessary. MAIN RESULTS We found 22 trials that met our inclusion criteria with a total of over 2310 participants (one study did not report number of participants). The included studies mostly had small numbers of participants (from 4 to 317) and relatively short follow-up periods (4 to 24 weeks). At baseline, six trials included only people with ulcers that were clinically infected; one trial included people with both infected and uninfected ulcers; two trials included people with non-infected ulcers; and the remaining 13 studies did not report infection status.Included studies employed various topical antimicrobial treatments, including antimicrobial dressings (e.g. silver, iodides), super-oxidised aqueous solutions, zinc hyaluronate, silver sulphadiazine, tretinoin, pexiganan cream, and chloramine. We performed the following five comparisons based on the included studies: Antimicrobial dressings compared with non-antimicrobial dressings: Pooled data from five trials with a total of 945 participants suggest (based on the average treatment effect from a random-effects model) that more wounds may heal when treated with an antimicrobial dressing than with a non-antimicrobial dressing: risk ratio (RR) 1.28, 95% confidence interval (CI) 1.12 to 1.45. These results correspond to an additional 119 healing events in the antimicrobial-dressing arm per 1000 participants (95% CI 51 to 191 more). We consider this low-certainty evidence (downgraded twice due to risk of bias). The evidence on adverse events or other outcomes was uncertain (very low-certainty evidence, frequently downgraded due to risk of bias and imprecision). Antimicrobial topical treatments (non dressings) compared with non-antimicrobial topical treatments (non dressings): There were four trials with a total of 132 participants in this comparison that contributed variously to the estimates of outcome data. Evidence was generally of low or very low certainty, and the 95% CIs spanned benefit and harm: proportion of wounds healed RR 2.82 (95% CI 0.56 to 14.23; 112 participants; 3 trials; very low-certainty evidence); achieving resolution of infection RR 1.16 (95% CI 0.54 to 2.51; 40 participants; 1 trial; low-certainty evidence); undergoing surgical resection RR 1.67 (95% CI 0.47 to 5.90; 40 participants; 1 trial; low-certainty evidence); and sustaining an adverse event (no events in either arm; 81 participants; 2 trials; very low-certainty evidence). Comparison of different topical antimicrobial treatments: We included eight studies with a total of 250 participants, but all of the comparisons were different and no data could be appropriately pooled. Reported outcome data were limited and we are uncertain about the relative effects of antimicrobial topical agents for each of our review outcomes for this comparison, that is wound healing, resolution of infection, surgical resection, and adverse events (all very low-certainty evidence). Topical antimicrobials compared with systemic antibiotics : We included four studies with a total of 937 participants. These studies reported no wound-healing data, and the evidence was uncertain for the relative effects on resolution of infection in infected ulcers and surgical resection (very low certainty). On average, there is probably little difference in the risk of adverse events between the compared topical antimicrobial and systemic antibiotics treatments: RR 0.91 (95% CI 0.78 to 1.06; moderate-certainty evidence - downgraded once for inconsistency). Topical antimicrobial agents compared with growth factor: We included one study with 40 participants. The only review-relevant outcome reported was number of ulcers healed, and these data were uncertain (very low-certainty evidence). AUTHORS' CONCLUSIONS The randomised controlled trial data on the effectiveness and safety of topical antimicrobial treatments for diabetic foot ulcers is limited by the availability of relatively few, mostly small, and often poorly designed trials. Based on our systematic review and analysis of the literature, we suggest that: 1) use of an antimicrobial dressing instead of a non-antimicrobial dressing may increase the number of diabetic foot ulcers healed over a medium-term follow-up period (low-certainty evidence); and 2) there is probably little difference in the risk of adverse events related to treatment between systemic antibiotics and topical antimicrobial treatments based on the available studies (moderate-certainty evidence). For each of the other outcomes we examined there were either no reported data or the available data left us uncertain as to whether or not there were any differences between the compared treatments. Given the high, and increasing, frequency of diabetic foot wounds, we encourage investigators to undertake properly designed randomised controlled trials in this area to evaluate the effects of topical antimicrobial treatments for both the prevention and the treatment of infection in these wounds and ultimately the effects on wound healing.
Collapse
Affiliation(s)
- Jo C Dumville
- University of Manchester, Manchester Academic Health Science CentreDivision of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine & HealthManchesterUKM13 9PL
| | - Benjamin A Lipsky
- Green Templeton College, University of OxfordDivision of Medical Sciences79 Stone MeadowOxfordUKOX2 6TD
| | - Christopher Hoey
- VA Puget Sound Health Care System Medical CenterPharmacy and Nutritional CareS‐Pharm‐1191660 S. Columbian WaySeattleWAUSA98108‐1597
| | - Mario Cruciani
- Azienda ULSS9 ScaligeraAntibiotic Stewardship ProgrammeVeronaItaly37135
| | - Marta Fiscon
- University of VeronaCentro Malattie DiffusiveULSS20VeronaItaly
| | - Jun Xia
- The Ingenuity Centre, The University of NottinghamSystematic Review Solutions LtdTriumph RoadNottinghamUKNG7 2TU
| | | |
Collapse
|
42
|
Zhang P, Wright JA, Osman AA, Nair SP. An aroD Ochre Mutation Results in a Staphylococcus aureus Small Colony Variant That Can Undergo Phenotypic Switching via Two Alternative Mechanisms. Front Microbiol 2017; 8:1001. [PMID: 28620368 PMCID: PMC5449664 DOI: 10.3389/fmicb.2017.01001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/18/2017] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus can undergo phenotypic switching between a normal colony phenotype (NCP) and a small colony variant (SCV). The SCV phenotype confers increased antibiotic resistance and the capacity to persist within human tissues and cells, and because these cells can revert back to the NCP they cause chronic and/or recurrent infections that are very difficult to treat. A complete picture of the genetic events that can lead to phenotypic switching in S. aureus is currently lacking. We describe the selection of an SCV with a previously unreported genetic alteration leading to an ochre mutation of aroD. In addition to the known mechanisms of phenotypic switching between the SCV and the NCP we describe a previously unreported mechanism involving tRNA ochre suppressors arising. The ochre suppressor strains had wild-type growth rates and restored antibiotic sensitivity, similar to the wild-type strain. However, whilst they had increased virulence compared to the SCV parent strain, their virulence was not restored to that of the NCP parental strain. These findings establish that phenotypic switching between the NCP and SCV states can give rise to strains with different pathogenic potential.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College LondonLondon, United Kingdom
| | - John A Wright
- Immunology Catalyst, GlaxoSmithKline plcStevenage, United Kingdom
| | - Ahmed A Osman
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College LondonLondon, United Kingdom
| | - Sean P Nair
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College LondonLondon, United Kingdom
| |
Collapse
|
43
|
Čepl J, Blahůšková A, Neubauer Z, Markoš A. Variations and heredity in bacterial colonies. Commun Integr Biol 2016; 9:e1261228. [PMID: 28042382 PMCID: PMC5193049 DOI: 10.1080/19420889.2016.1261228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 01/24/2023] Open
Abstract
Spontaneous variation in appearance was studied in bacterial colonies of Serratia marcescens F morphotype1: (i) A defined array of non-heritable phenotype variations does appear repeatedly; (ii) The presence of colonies of different bacterial species will narrow the variability toward the typical F appearance, as if such an added environmental factor curtailed the capacity of colony morphospace; (iii) Similarly the morphospace becomes reduced by random mutations leading to new, heritable morphotypes—at the same time opening a new array of variations typical for the mutant but not accessible directly from the original F morphospace. Results are discussed in context with biphasic model of early morphogenesis applicable to all multicellular bodies.
Collapse
Affiliation(s)
- Jaroslav Čepl
- Czech University of Life Sciences, Faculty of Forestry and Wood Sciences , Prague, Czechia
| | - Anna Blahůšková
- Charles University in Prague, Faculty of Sciences , Prague, Czechia
| | - Zdeněk Neubauer
- Charles University in Prague, Faculty of Sciences , Prague, Czechia
| | - Anton Markoš
- Charles University in Prague, Faculty of Sciences , Prague, Czechia
| |
Collapse
|
44
|
Clinical Significance and Pathogenesis of Staphylococcal Small Colony Variants in Persistent Infections. Clin Microbiol Rev 2016; 29:401-27. [PMID: 26960941 DOI: 10.1128/cmr.00069-15] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Small colony variants (SCVs) were first described more than 100 years ago for Staphylococcus aureus and various coagulase-negative staphylococci. Two decades ago, an association between chronic staphylococcal infections and the presence of SCVs was observed. Since then, many clinical studies and observations have been published which tie recurrent, persistent staphylococcal infections, including device-associated infections, bone and tissue infections, and airway infections of cystic fibrosis patients, to this special phenotype. By their intracellular lifestyle, SCVs exhibit so-called phenotypic (or functional) resistance beyond the classical resistance mechanisms, and they can often be retrieved from therapy-refractory courses of infection. In this review, the various clinical infections where SCVs can be expected and isolated, diagnostic procedures for optimized species confirmation, and the pathogenesis of SCVs, including defined underlying molecular mechanisms and the phenotype switch phenomenon, are presented. Moreover, relevant animal models and suggested treatment regimens, as well as the requirements for future research areas, are highlighted.
Collapse
|
45
|
Kluin OS, Busscher HJ, Neut D, van der Mei HC. Poly(trimethylene carbonate) as a carrier for rifampicin and vancomycin to target therapy-recalcitrant staphylococcal biofilms. J Orthop Res 2016; 34:1828-1837. [PMID: 26876651 DOI: 10.1002/jor.23194] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/08/2016] [Indexed: 02/04/2023]
Abstract
Standard antibiotic therapy in osteomyelitis patients is of limited value when methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus epidermidis (MRSE), or small-colony variants (SCV) are present. Far better results are obtained by local drug delivery of antibiotic combinations including rifampicin, using a suitable carrier. We therefore investigated release kinetics of antibiotics from biodegradable poly(trimethylene carbonate) (PTMC) and in vitro biofilm inhibition of MRSA, MRSE, and S. aureus SCV strains in the course of 24, 72, and 168 h treatment by PTMC, either unloaded, gentamicin-loaded, loaded with rifampicin and fosfomycin, or rifampicin and vancomycin. PTMC appeared to be a suitable carrier for rifampicin alone or in combination with other antibiotics. Biofilm colony forming units and metabolic activity measurement (MTT assay) demonstrated significant (p < 0.05) inhibition for all strains when PTMC loaded with rifampicin and vancomycin was employed, especially after 168 h treatment. Confocal laser scanning microscopy images showed similar qualitative results. PTMC loaded with only gentamicin did not show any inhibition. This exemplifies that PTMC loaded with rifampicin and vancomycin holds promise for the treatment of recalcitrant osteomyelitis. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1828-1837, 2016.
Collapse
Affiliation(s)
- Otto S Kluin
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands
| | - Henk J Busscher
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands
| | - Daniëlle Neut
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands.
| | - Henny C van der Mei
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands
| |
Collapse
|
46
|
Sulabh S, Bhushan B, Panigrahi M, Verma A, Baba NA, Kumar P. Differential response of immune-related genes to peptidoglycan and lipoteichoic acid challenge in vitro. Vet World 2016; 9:983-988. [PMID: 27733800 PMCID: PMC5057038 DOI: 10.14202/vetworld.2016.983-988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/01/2016] [Indexed: 01/27/2023] Open
Abstract
AIM To study the effect of Staphylococcus aureus cell wall antigens, peptidoglycan (PGN) and lipoteichoic acid (LTA) challenge on immune cells present in bovine peripheral blood mononuclear cells (PBMCs). MATERIALS AND METHODS In this study, efforts have been made to investigate the effects of three combinations (10+10, 20+20 and 30+30 μg/ml) of PGN and LTA obtained from S. aureus. These antigens were used to challenge the bovine PBMCs. After 6 h of incubation quantitative, real time-polymerase chain reaction was used to study toll-like receptor 2 (TLR-2) and major cytokine mRNA expression in bovine PBMC challenged with three different antigen blends. RESULTS The results indicated that mRNA level of interferon gamma is influenced by the expression of TLR-2 gene. Tumor necrosis factor-alpha (TNF-α), interleukin 10 (IL-10), and IL-8 genes showed a maximum response at a dose of 10 μg of PGN and 10 μg of LTA challenge per ml of culture medium. The outcome also suggests that both IL-10 and IL-8 followed the expression pattern of TNF-α. CONCLUSION A dose of 10 μg of PGN and 10 μg of LTA per ml of culture medium was found to be most suitable for challenging PBMC.
Collapse
Affiliation(s)
- Sourabh Sulabh
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly - 243 122, Uttar Pradesh, India
| | - Bharat Bhushan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly - 243 122, Uttar Pradesh, India
| | - Manjit Panigrahi
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly - 243 122, Uttar Pradesh, India
| | - Ankita Verma
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly - 243 122, Uttar Pradesh, India
| | - Naseer Ahmad Baba
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly - 243 122, Uttar Pradesh, India
| | - Pushpendra Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly - 243 122, Uttar Pradesh, India
| |
Collapse
|
47
|
Alreshidi MM, Dunstan RH, Gottfries J, Macdonald MM, Crompton MJ, Ang CS, Williamson NA, Roberts TK. Changes in the Cytoplasmic Composition of Amino Acids and Proteins Observed in Staphylococcus aureus during Growth under Variable Growth Conditions Representative of the Human Wound Site. PLoS One 2016; 11:e0159662. [PMID: 27442022 PMCID: PMC4956324 DOI: 10.1371/journal.pone.0159662] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/05/2016] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen responsible for a high proportion of nosocomial infections. This study was conducted to assess the bacterial responses in the cytoplasmic composition of amino acids and ribosomal proteins under various environmental conditions designed to mimic those on the human skin or within a wound site: pH6-8, temperature 35–37°C, and additional 0–5% NaCl. It was found that each set of environmental conditions elicited substantial adjustments in cytoplasmic levels of glutamic acid, aspartic acid, proline, alanine and glycine (P< 0.05). These alterations generated characteristic amino acid profiles assessed by principle component analysis (PCA). Substantial alterations in cytoplasmic amino acid and protein composition occurred during growth under conditions of higher salinity stress implemented via additional levels of NaCl in the growth medium. The cells responded to additional NaCl at pH 6 by reducing levels of ribosomal proteins, whereas at pH 8 there was an upregulation of ribosomal proteins compared with the reference control. The levels of two ribosomal proteins, L32 and S19, remained constant across all experimental conditions. The data supported the hypothesis that the bacterium was continually responding to the dynamic environment by modifying the proteome and optimising metabolic homeostasis.
Collapse
Affiliation(s)
- Mousa M. Alreshidi
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
- Department of Biology, College of Science, University of Ha’il, P.O. 2440, Hail, Saudi Arabia
| | - R. Hugh Dunstan
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
- * E-mail:
| | - Johan Gottfries
- Department of Chemistry and Molecular Biology, Gothenburg University, Sweden
| | - Margaret M. Macdonald
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| | - Marcus J. Crompton
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria 3010, Australia
| | - Nicholas A. Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria 3010, Australia
| | - Tim K. Roberts
- Metabolic Research Group, Faculty of Science and Information Technology, School of Environmental and Life Sciences, Department of Biology, University Drive, Callaghan, 2308, NSW, Australia
| |
Collapse
|
48
|
Oyama T, Miyazaki M, Yoshimura M, Takata T, Ohjimi H, Jimi S. Biofilm-Forming Methicillin-Resistant Staphylococcus aureus Survive in Kupffer Cells and Exhibit High Virulence in Mice. Toxins (Basel) 2016; 8:toxins8070198. [PMID: 27376326 PMCID: PMC4963831 DOI: 10.3390/toxins8070198] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/15/2016] [Accepted: 06/24/2016] [Indexed: 11/26/2022] Open
Abstract
Although Staphylococcus aureus is part of the normal body flora, heavy usage of antibiotics has resulted in the emergence of methicillin-resistant strains (MRSA). MRSA can form biofilms and cause indwelling foreign body infections, bacteremia, soft tissue infections, endocarditis, and osteomyelitis. Using an in vitro assay, we screened 173 clinical blood isolates of MRSA and selected 20 high-biofilm formers (H-BF) and low-biofilm formers (L-BF). These were intravenously administered to mice and the general condition of mice, the distribution of bacteria, and biofilm in the liver, lung, spleen, and kidney were investigated. MRSA count was the highest in the liver, especially within Kupffer cells, which were positive for acid polysaccharides that are associated with intracellular biofilm. After 24 h, the general condition of the mice worsened significantly in the H-BF group. In the liver, bacterial deposition and aggregation and the biofilm-forming spot number were all significantly greater for H-BF group than for L-BF. CFU analysis revealed that bacteria in the H-BF group survived for long periods in the liver. These results indicate that the biofilm-forming ability of MRSA is a crucial factor for intracellular persistence, which could lead to chronic infections.
Collapse
Affiliation(s)
- Takuto Oyama
- Department of Plastic, Reconstructive and Aesthetic Surgery, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Motoyasu Miyazaki
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Chikusino 818-8502, Japan.
| | - Michinobu Yoshimura
- Department of Medical Oncology, Hematology, and Infectious Diseases, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Tohru Takata
- Department of Medical Oncology, Hematology, and Infectious Diseases, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Hiroyuki Ohjimi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Shiro Jimi
- Central Laboratory for Pathology and Morphology, Department of Pathology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| |
Collapse
|
49
|
Li W, Li Y, Wu Y, Cui Y, Liu Y, Shi X, Zhang Q, Chen Q, Sun Q, Hu Q. Phenotypic and genetic changes in the life cycle of small colony variants of Salmonella enterica serotype Typhimurium induced by streptomycin. Ann Clin Microbiol Antimicrob 2016; 15:37. [PMID: 27245674 PMCID: PMC4888536 DOI: 10.1186/s12941-016-0151-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/18/2016] [Indexed: 01/29/2023] Open
Abstract
Background Small colony variants (SCVs), constituting a slow-growing subpopulation of bacteria that facilitates persistence in lethal environmental conditions, are able to revert to the phenotype of rapid growth for further proliferation and transmission. Salmonella enterica serotype Typhimurium is one of the most important foodborne pathogens. This study investigated the genetic mechanisms how SCVs induced by streptomycin reverted to the fast-growing phenotype and the phenotypic changes of SCVs among their complete life cycle in S.Typhimurium. Methods Salmonella Typhimurium SCVs were obtained by streptomycin treatment and their revertants were collected in the absence of antibiotics. The fitness, antimicrobial susceptibility, biofilm formation, and the biofilm-related genes expression were analyzed in comparison to their wild type strain, and the whole genome sequencing was performed to identify the genetic changes in the life cycle of S. Typhimurium SCVs. Results Small colony variants were characterized by an increased antimicrobial resistance to streptomycin (64-fold), imipenem (twofold), and gentamicin (fourfold). A significant increase in biofilm production with higher expression of csgB was observed in SCVs (P < 0.01). The genetic alterations of all SCVs occurred in ubiE gene (coenzyme Q8 and menaquinone synthesis) with frameshift mutations. However, all fast-growing revertants again lost the trait of increased biofilm production (P > 0.05), in which two modes of the genetic changes for reversing to the rapidly growing form were observed: four revertants harbored a secondary mutation in ubiE, which reinstated most of the amino acid sequence of the ubiE, and other four revertants harbored a mutation in prfB. Conclusions Salmonella Typhimurium could switch to the phenotype of SCVs under the treatment of streptomycin by a mutation in ubiE, partially combined with increased production of biofilm, and these SCVs could escape from growth restriction by a compensatory mutation in prfB or a new mutation in ubiE. These findings may contribute to establishing phenotype-directed treatments against SCVs of S.Typhimurium. Electronic supplementary material The online version of this article (doi:10.1186/s12941-016-0151-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wanli Li
- Key Laboratory of Bio-resources and Eco-environment of the Ministry of Education, College of Life Sciences, Sichuan University, 29# Wangjiang Rd., Chengdu, Sichuan, 610064, P. R. China
| | - Yinghui Li
- Shenzhen Major Infectious Disease Control Key Laboratory, Shenzhen Centre for Disease Control and Prevention, Shenzhen, Guangdong, 518055, P. R. China
| | - Yarong Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, P. R. China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, P. R. China
| | - Yao Liu
- Key Laboratory of Bio-resources and Eco-environment of the Ministry of Education, College of Life Sciences, Sichuan University, 29# Wangjiang Rd., Chengdu, Sichuan, 610064, P. R. China
| | - Xiaolu Shi
- Shenzhen Major Infectious Disease Control Key Laboratory, Shenzhen Centre for Disease Control and Prevention, Shenzhen, Guangdong, 518055, P. R. China
| | - Qian Zhang
- Shenzhen Major Infectious Disease Control Key Laboratory, Shenzhen Centre for Disease Control and Prevention, Shenzhen, Guangdong, 518055, P. R. China
| | - Qiongcheng Chen
- Shenzhen Major Infectious Disease Control Key Laboratory, Shenzhen Centre for Disease Control and Prevention, Shenzhen, Guangdong, 518055, P. R. China
| | - Qun Sun
- Key Laboratory of Bio-resources and Eco-environment of the Ministry of Education, College of Life Sciences, Sichuan University, 29# Wangjiang Rd., Chengdu, Sichuan, 610064, P. R. China.
| | - Qinghua Hu
- Shenzhen Major Infectious Disease Control Key Laboratory, Shenzhen Centre for Disease Control and Prevention, Shenzhen, Guangdong, 518055, P. R. China.
| |
Collapse
|
50
|
Dayan GH, Mohamed N, Scully IL, Cooper D, Begier E, Eiden J, Jansen KU, Gurtman A, Anderson AS. Staphylococcus aureus: the current state of disease, pathophysiology and strategies for prevention. Expert Rev Vaccines 2016; 15:1373-1392. [PMID: 27118628 DOI: 10.1080/14760584.2016.1179583] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is both a commensal organism and also an important opportunistic human pathogen, causing a variety of community and hospital-associated pathologies, such as bacteremia-sepsis, endocarditis, pneumonia, osteomyelitis, arthritis and skin diseases. The resurgence of S. aureus during the last decade in many settings has been facilitated not only by bacterial antibiotic resistance mechanisms but also by the emergence of new S. aureus clonal types with increased expression of virulence factors and the capacity to neutralize the host immune response. Prevention of the spread of S. aureus infection relies on the use of contact precautions and adequate procedures for infection control that so far have not been fully effective. Prevention using a prophylactic vaccine would complement these processes, having the potential to bring additional, significant progress toward decreasing invasive disease due to S. aureus.
Collapse
Affiliation(s)
- Gustavo H Dayan
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Naglaa Mohamed
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Ingrid L Scully
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - David Cooper
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Elizabeth Begier
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Joseph Eiden
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Kathrin U Jansen
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | | | | |
Collapse
|