201
|
Tkhilaishvili T, Wang L, Tavanti A, Trampuz A, Di Luca M. Antibacterial Efficacy of Two Commercially Available Bacteriophage Formulations, Staphylococcal Bacteriophage and PYO Bacteriophage, Against Methicillin-Resistant Staphylococcus aureus: Prevention and Eradication of Biofilm Formation and Control of a Systemic Infection of Galleria mellonella Larvae. Front Microbiol 2020; 11:110. [PMID: 32117136 PMCID: PMC7018685 DOI: 10.3389/fmicb.2020.00110] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/17/2020] [Indexed: 12/16/2022] Open
Abstract
Sessile bacteria growing on surfaces are more resistant to standard antibiotics than their planktonic counterpart. Due to their antimicrobial properties, bacteriophages have re-emerged as a promising approach to treat bacterial biofilm-associated infections. Here, we evaluated the ability of two commercially available phage formulations, Staphylococcal bacteriophage (containing the monophage Sb-1) and PYO bacteriophage (a polyphage), in preventing and eradicating an in vitro biofilm of methicillin-resistant Staphylococcus aureus (MRSA) by isothermal microcalorimetry and high-resolution confocal laser scanning microscopy (CLSM). Moreover, to assess the potential in vivo efficacy of both phage preparations, a Galleria mellonella model of MRSA systemic infection was used. Microcalorimetry measurement showed that 107 PFU/ml (the highest tested titer) of both phage formulations were able to inhibit planktonic growth in a concentration-dependent manner. However, MRSA biofilm was eradicated only by co-incubation of 5–7 days with the highest phage titers, respectively. In the experiments of biofilm prevention, isothermal microcalorimetry revealed that the heat production was completely abolished in the presence of sub-inhibitory titers (104 PFU/ml) of phages. These data were also confirmed by confocal laser scanning microscopy. Both phage formulations increased the survival of G. mellonella larvae preventing or treating MRSA infection compared to untreated control. In conclusion, tested phage formulations are promising for preventing device colonization and killing biofilm bacteria attached on a surface. Novel strategies for direct coating and release of phages from material should be investigated.
Collapse
Affiliation(s)
- Tamta Tkhilaishvili
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lei Wang
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Andrej Trampuz
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mariagrazia Di Luca
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
202
|
Khan F, Lee JW, Pham DTN, Kim YM. Chitooligosaccharides as Antibacterial, Antibiofilm, Antihemolytic and Anti-Virulence Agent against Staphylococcus aureus. Curr Pharm Biotechnol 2020; 20:1223-1233. [PMID: 31475895 DOI: 10.2174/1389201020666190902130722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Staphylococcus aureus nosocomial infections with a high mortality rate in human and animals have been reported to associate with bacterial biofilm formation, along with the secretion of numerous virulence factors. Therefore, the inhibition of biofilm formation and attenuation of virulence determinants are considered as a promising solution to combat the spread of S. aureus infections. Modern trends in antibiofilm therapies have opted for the active agents that are biocompatible, biodegradable, non-toxic and cost-effective. Owning the aforementioned properties, chitosan, a natural N-acetylated carbohydrate biopolymer derived from chitin, has been favorably employed. Recently, the chitosan structure has been chemically modified into Chitooligosaccharides (COS) to overcome its limited solubility in water, thus widening chitosan applications in modern antibiofilm research. In the present study, we have investigated the antibacterial, antibiofilm and anti-virulence activities against S. aureus of COS of different molecular weights dissolved in neutral water. METHODS The study of bactericidal activity was performed using the micro-dilution method while the biofilm inhibition assay was performed using crystal-violet staining method and confirmed by scanning electron microscopic analysis. The inhibition of amyloid protein production was confirmed by Congo Red staining. RESULTS Results showed that low molecular weight COS exhibited bactericidal activity and reduced the bacterial amylogenesis, hemolytic activity as well as H2O2 resistance properties, while slightly inhibiting biofilm formation. The present study provides a new insight for further applications of the water-soluble COS as a safe and cost-effective drug for the treatment of S. aureus biofilm-associated infections. CONCLUSION Reducing the molecular weight of chitosan in the form of COS has become an effective strategy to maintain chitosan biological activity while improving its water solubility. The low molecular weight COS investigated in this study have effectively performed antibacterial, antibiofilm and antivirulence properties against S. aureus.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea
| | - Jang-Won Lee
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| | - Dung T N Pham
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| | - Young-Mog Kim
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea.,Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
203
|
Epstein IJ, Rosenberg E, Stuber R, Choi MB, Donnenfeld ED, Perry HD. Double-Masked and Unmasked Prospective Study of Terpinen-4-ol Lid Scrubs With Microblepharoexfoliation for the Treatment of Demodex Blepharitis. Cornea 2020; 39:408-416. [DOI: 10.1097/ico.0000000000002243] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
204
|
Shokeen B, Park J, Duong E, Rambhia S, Paul M, Weinberg A, Shi W, Lux R. Role of FAD-I in Fusobacterial Interspecies Interaction and Biofilm Formation. Microorganisms 2020; 8:E70. [PMID: 31906541 PMCID: PMC7023056 DOI: 10.3390/microorganisms8010070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/25/2019] [Accepted: 12/31/2019] [Indexed: 11/17/2022] Open
Abstract
: RadD, a major adhesin of oral fusobacteria, is part of a four-gene operon encoding the small lipoprotein FAD-I and two currently uncharacterized small proteins encoded by the rapA and rapB genes. Previously, we described a role for FAD-I in the induction of human B-defensin 2 (hBD2) upon contact with oral epithelial cells. Here, we investigated potential roles for fad-I, rapA, and rapB in interspecies interaction and biofilm formation. Gene inactivation mutants were generated for each of these genes in the nucleatum and polymorphum subspecies of Fusobacterium nucleatum and characterized for their adherence to partner species, biofilm formation, and operon transcription. Binding to Streptococcus gordonii was increased in all mutant strains with Δfad-I having the most significant effect. This increased adherence was directly proportional to elevated radD transcript levels and resulted in significantly different architecture and height of the biofilms formed by Δfad-I and S. gordonii compared to the wild-type parent. In conclusion, FAD-I is important for fusobacterial interspecies interaction as its lack leads to increased production of the RadD adhesin suggesting a role of FAD-I in its regulation. This regulatory effect does not require the presence of functional RadD.
Collapse
Affiliation(s)
- Bhumika Shokeen
- Section of Periodontics, Division of Constitutive & Regenerative Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Jane Park
- Section of Periodontics, Division of Constitutive & Regenerative Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Emily Duong
- Section of Periodontics, Division of Constitutive & Regenerative Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Sonam Rambhia
- Section of Periodontics, Division of Constitutive & Regenerative Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Manash Paul
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Aaron Weinberg
- Department of Biological Sciences, Case Western Reserve University, Cleveland, OH 44106-4905, USA
| | - Wenyuan Shi
- The Forsyth Institute, Cambridge, MA 02142, USA
| | - Renate Lux
- Section of Periodontics, Division of Constitutive & Regenerative Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| |
Collapse
|
205
|
Verderosa AD, Dhouib R, Fairfull-Smith KE, Totsika M. Nitroxide Functionalized Antibiotics Are Promising Eradication Agents against Staphylococcus aureus Biofilms. Antimicrob Agents Chemother 2019; 64:e01685-19. [PMID: 31636066 PMCID: PMC7187575 DOI: 10.1128/aac.01685-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/09/2019] [Indexed: 01/10/2023] Open
Abstract
Treatment of biofilm-related Staphylococcus aureus infections represents an important medical challenge worldwide, as biofilms, even those involving drug-susceptible S. aureus strains, are highly refractory to conventional antibiotic therapy. Nitroxides were recently shown to induce the dispersal of Gram-negative biofilms in vitro, but their action against Gram-positive bacterial biofilms remains unknown. Here, we demonstrate that the biofilm dispersal activity of nitroxides extends to S. aureus, a clinically important Gram-positive pathogen. Coadministration of the nitroxide CTEMPO (4-carboxy-2,2,6,6-tetramethylpiperidin-1-yloxyl) with ciprofloxacin significantly improved the biofilm eradication activity of the antibiotic against S. aureus Moreover, covalently linking the nitroxide to the antibiotic moiety further reduced the ciprofloxacin minimal biofilm eradication concentration. Microscopy analysis revealed that fluorescent nitroxide-antibiotic hybrids could penetrate S. aureus biofilms and enter cells localized at the surface and base of the biofilm structure. No toxicity to human cells was observed for the nitroxide CTEMPO or the nitroxide-antibiotic hybrids. Taken together, our results show that nitroxides can mediate the dispersal of Gram-positive biofilms and that dual-acting biofilm eradication antibiotics may provide broad-spectrum therapies for the treatment of biofilm-related infections.
Collapse
Affiliation(s)
- Anthony D Verderosa
- School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Rabeb Dhouib
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Makrina Totsika
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
206
|
Hannachi N, Baudoin JP, Prasanth A, Habib G, Camoin-Jau L. The distinct effects of aspirin on platelet aggregation induced by infectious bacteria. Platelets 2019; 31:1028-1038. [PMID: 31856631 DOI: 10.1080/09537104.2019.1704717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bacteria induce platelet aggregation triggered by several mechanisms. The goal of this work was to characterize platelet aggregates induced by different bacterial strains and to quantify the effect of aspirin treatment using aggregation tests, as well as a novel approach based on confocal analysis. Blood samples were obtained from either healthy donors (n = 27) or patients treated with long-term aspirin (n = 15). The bacterial species included were Staphylococcus aureus, Enterococcus faecalis, and Streptococcus sanguinis. The different aggregate's ultrastructures depending on the bacterial strain were analyzed using Scanning electron microscopy. Quantification of the size of the platelet aggregates, their mean number as well as the bacterial impregnation within the aggregates was performed using confocal laser scanning light microscopy. Light Transmission Aggregometry was also performed. Our results reported distinct characteristics of platelet aggregates depending on the bacterial strain. Using confocal analysis, we have shown that aspirin significantly reduced platelet aggregation induced by S. aureus (p = .003) and E. faecalis (p = .006) with no effect in the case of S. sanguinis (p = .529). The results of the aggregometry were concordant with those of the confocal technique in the case of S. aureus and S. sanguinis. Interestingly, aggregation induced by E. faecalis was detected only with confocal analysis. In conclusion, our confocal scanning microscopy allowed a detailed study of the platelet aggregation induced by bacteria. We showed that aspirin acts on bacterial-induced platelet aggregation depending on the species. These results are in favor of the use of aspirin considering the species and the bacterial strain involved.
Collapse
Affiliation(s)
- Nadji Hannachi
- Département d'infectiologie, MEPHI, IHU Méditerranée infection, Aix Marseille Univ, IRD, AP-HM , Marseille, France
| | - Jean-Pierre Baudoin
- Département d'infectiologie, MEPHI, IHU Méditerranée infection, Aix Marseille Univ, IRD, AP-HM , Marseille, France
| | - Arsha Prasanth
- Département d'infectiologie, MEPHI, IHU Méditerranée infection, Aix Marseille Univ, IRD, AP-HM , Marseille, France
| | - Gilbert Habib
- Département d'infectiologie, MEPHI, IHU Méditerranée infection, Aix Marseille Univ, IRD, AP-HM , Marseille, France.,Département de cardiologie, la Timone Hospital, AP-HM , Marseille, France
| | - Laurence Camoin-Jau
- Département d'infectiologie, MEPHI, IHU Méditerranée infection, Aix Marseille Univ, IRD, AP-HM , Marseille, France.,Laboratoire d'Hématologie, La Timone Hospital, APHM , Marseille, France
| |
Collapse
|
207
|
Wang J, Jiao H, Meng J, Qiao M, Du H, He M, Ming K, Liu J, Wang D, Wu Y. Baicalin Inhibits Biofilm Formation and the Quorum-Sensing System by Regulating the MsrA Drug Efflux Pump in Staphylococcus saprophyticus. Front Microbiol 2019; 10:2800. [PMID: 31921008 PMCID: PMC6915091 DOI: 10.3389/fmicb.2019.02800] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/18/2019] [Indexed: 11/29/2022] Open
Abstract
Staphylococcus saprophyticus (S. saprophyticus) is one of the main pathogens that cause serious infection due to its acquisition of antibiotic resistance. The efflux pump decreases antibiotic abundance, and biofilm compromises the penetration of antibiotics. It has been reported that baicalin is a potential agent to inhibit efflux pumps, biofilm formation, and quorum-sensing systems. The purpose of this study was to investigate whether baicalin can inhibit S. saprophyticus biofilm formation and the quorum-sensing system by inhibiting the MsrA efflux pump. First, the mechanism of baicalin inhibiting efflux was investigated by the ethidium bromide (EtBr) efflux assay, measurement of ATP content, and pyruvate kinase (PK) activities. These results revealed that baicalin significantly reduced the efflux of EtBr, the ATP content, and the activity of PK. Moreover, its role in biofilm formation and the agr system was studied by crystal violet staining, confocal laser scanning microscopy, scanning electron microscopy, and real-time polymerase chain reaction. These results showed that baicalin decreased biofilm formation, inhibited bacterial aggregation, and downregulated mRNA transcription levels of the quorum-sensing system regulators agrA, agrC, RNAIII, and sarA. Correlation analysis indicated that there was a strong positive correlation between the efflux pump and biofilm formation and the agr system. We demonstrate for the first time that baicalin inhibits biofilm formation and the agr quorum-sensing system by inhibiting the efflux pump in S. saprophyticus. Therefore, baicalin is a potential therapeutic agent for S. saprophyticus biofilm-associated infections.
Collapse
Affiliation(s)
- Jinli Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Haihong Jiao
- Key Laboratory of Tarim Animal Husbandry Science and Technology of Xinjiang Production & Construction Corps, College of Animal Science, Tarim University, Alar, China
| | - Jinwu Meng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Mingyu Qiao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongxu Du
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Miao He
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ke Ming
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiaguo Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Deyun Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yi Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
208
|
Sub-Inhibitory Concentrations of Sodium Houttuyfonate in Combination with Erythromycin Inhibit Biofilm Formation and Expression of IcaA in Staphylococcus epidermidis. Jundishapur J Microbiol 2019. [DOI: 10.5812/jjm.98009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
209
|
Huigens RW, Abouelhassan Y, Yang H. Phenazine Antibiotic-Inspired Discovery of Bacterial Biofilm-Eradicating Agents. Chembiochem 2019; 20:2885-2902. [PMID: 30811834 PMCID: PMC7325843 DOI: 10.1002/cbic.201900116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Indexed: 12/19/2022]
Abstract
Bacterial biofilms are surface-attached communities of slow-growing and non-replicating persister cells that demonstrate high levels of antibiotic tolerance. Biofilms occur in nearly 80 % of infections and present unique challenges to our current arsenal of antibiotic therapies, all of which were initially discovered for their abilities to target rapidly dividing, free-floating planktonic bacteria. Bacterial biofilms are credited as the underlying cause of chronic and recurring bacterial infections. Innovative approaches are required to identify new small molecules that operate through bacterial growth-independent mechanisms to effectively eradicate biofilms. One source of inspiration comes from within the lungs of young cystic fibrosis (CF) patients, who often endure persistent Staphylococcus aureus infections. As these CF patients age, Pseudomonas aeruginosa co-infects the lungs and utilizes phenazine antibiotics to eradicate the established S. aureus infection. Our group has taken a special interest in this microbial competition strategy and we are investigating the potential of phenazine antibiotic-inspired compounds and synthetic analogues thereof to eradicate persistent bacterial biofilms. To discover new biofilm-eradicating agents, we have established an interdisciplinary research program involving synthetic medicinal chemistry, microbiology and molecular biology. From these efforts, we have identified a series of halogenated phenazines (HPs) that potently eradicate bacterial biofilms, and future work aims to translate these preliminary findings into ground-breaking clinical advances for the treatment of persistent biofilm infections.
Collapse
Affiliation(s)
- Robert W. Huigens
- Department of Medicinal Chemistry; Center for Natural Products Drug Discovery and Development (CNPD3); University of Florida, Gainesville, FL, USA
| | - Yasmeen Abouelhassan
- Department of Medicinal Chemistry; Center for Natural Products Drug Discovery and Development (CNPD3); University of Florida, Gainesville, FL, USA
| | - Hongfen Yang
- Department of Medicinal Chemistry; Center for Natural Products Drug Discovery and Development (CNPD3); University of Florida, Gainesville, FL, USA
| |
Collapse
|
210
|
Muthukrishnan G, Masters EA, Daiss JL, Schwarz EM. Mechanisms of Immune Evasion and Bone Tissue Colonization That Make Staphylococcus aureus the Primary Pathogen in Osteomyelitis. Curr Osteoporos Rep 2019; 17:395-404. [PMID: 31721069 PMCID: PMC7344867 DOI: 10.1007/s11914-019-00548-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Staphylococcus aureus is the primary pathogen responsible for osteomyelitis, which remains a major healthcare burden. To understand its dominance, here we review the unique pathogenic mechanisms utilized by S. aureus that enable it to cause incurable osteomyelitis. RECENT FINDINGS Using an arsenal of toxins and virulence proteins, S. aureus kills and usurps immune cells during infection, to produce non-neutralizing pathogenic antibodies that thwart adaptive immunity. S. aureus also has specific mechanisms for distinct biofilm formation on implants, necrotic bone tissue, bone marrow, and within the osteocyte lacuno-canicular networks (OLCN) of live bone. In vitro studies have also demonstrated potential for intracellular colonization of osteocytes, osteoblasts, and osteoclasts. S. aureus has evolved a multitude of virulence mechanisms to achieve life-long infection of the bone, most notably colonization of OLCN. Targeting S. aureus proteins involved in these pathways could provide new targets for antibiotics and immunotherapies.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
211
|
LIU QING, CHEN XINTONG, LI XIAOYA, HONG JIANPING, JIANG GUIXIAN, LIANG HONGYU, LIU WENWEN, XU ZHENG, ZHANG JING, WANG WEI, XIAO LIANG. The Diversity of the Endobiotic Bacterial Communities in the Four Jellyfish Species. Pol J Microbiol 2019; 68:465-476. [PMID: 31880891 PMCID: PMC7260641 DOI: 10.33073/pjm-2019-046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/04/2019] [Accepted: 09/19/2019] [Indexed: 02/01/2023] Open
Abstract
The associated microbiota plays an essential role in the life process of jellyfish. The endobiotic bacterial communities from four common jellyfish Phyllorhiza punctata, Cyanea capillata, Chrysaora melanaster, and Aurelia coerulea were comparatively analyzed by 16S rDNA sequencing in this study. Several 1049 OTUs were harvested from a total of 130 183 reads. Tenericutes (68.4%) and Firmicutes (82.1%) are the most abundant phyla in P. punctata and C. melanaster, whereas C. capillata and A. coerulea share the same top phylum Proteobacteria (76.9% vs. 78.3%). The classified OTUs and bacterial abundance greatly decrease from the phylum to genus level. The top 20 matched genera only account for 9.03% of the total community in P. punctata, 48.9% in C. capillata, 83.05% in C. melanaster, and 58.1% in A. coerulea, respectively. The heatmap of the top 50 genera shows that the relative abundances in A. coerulea and C. capillata are far richer than that in P. punctata and C. melanaster. Moreover, a total of 41 predictive functional categories at KEGG level 2 were identified. Our study indicates the independent diversity of the bacterial communities in the four common Scyphomedusae, which might involve in the metabolism and environmental information processing of the hosts. The associated microbiota plays an essential role in the life process of jellyfish. The endobiotic bacterial communities from four common jellyfish Phyllorhiza punctata, Cyanea capillata, Chrysaora melanaster, and Aurelia coerulea were comparatively analyzed by 16S rDNA sequencing in this study. Several 1049 OTUs were harvested from a total of 130 183 reads. Tenericutes (68.4%) and Firmicutes (82.1%) are the most abundant phyla in P. punctata and C. melanaster, whereas C. capillata and A. coerulea share the same top phylum Proteobacteria (76.9% vs. 78.3%). The classified OTUs and bacterial abundance greatly decrease from the phylum to genus level. The top 20 matched genera only account for 9.03% of the total community in P. punctata, 48.9% in C. capillata, 83.05% in C. melanaster, and 58.1% in A. coerulea, respectively. The heatmap of the top 50 genera shows that the relative abundances in A. coerulea and C. capillata are far richer than that in P. punctata and C. melanaster. Moreover, a total of 41 predictive functional categories at KEGG level 2 were identified. Our study indicates the independent diversity of the bacterial communities in the four common Scyphomedusae, which might involve in the metabolism and environmental information processing of the hosts.
Collapse
Affiliation(s)
- QING LIU
- College of Animal Science and Veterinary Medicine; ShanXi Agricultural University, TaiGu, ShanXi, China
| | - XINTONG CHEN
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Resources and Environment; ShanXi Agricultural University, ShanXi, TaiGu, China
| | - XIAOYA LI
- College of Animal Science and Veterinary Medicine; ShanXi Agricultural University, TaiGu, ShanXi, China
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - JIANPING HONG
- College of Resources and Environment; ShanXi Agricultural University, ShanXi, TaiGu, China
| | - GUIXIAN JIANG
- Clinical Medicine, Grade 2015, Second Military Medical University (Naval Medical University), Shanghai, China
| | - HONGYU LIANG
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Traditional Chinese Medicine; Jilin Agricultural University, Changchun, Jilin, China
| | - WENWEN LIU
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Traditional Chinese Medicine; Jilin Agricultural University, Changchun, Jilin, China
| | - ZHENG XU
- Administration Office for Scientific Research, Second Military Medical University (Naval Medical University), Shanghai, China
| | - JING ZHANG
- College of Traditional Chinese Medicine; Jilin Agricultural University, Changchun, Jilin, China
| | - WEI WANG
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - LIANG XIAO
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| |
Collapse
|
212
|
Anti-biofilm Effect of β-Lapachone and Lapachol Oxime Against Isolates of Staphylococcus aureus. Curr Microbiol 2019; 77:204-209. [DOI: 10.1007/s00284-019-01818-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/18/2019] [Indexed: 10/25/2022]
|
213
|
Rahim MI, Szafrański SP, Ingendoh-Tsakmakidis A, Stiesch M, Mueller PP. Evidence for inoculum size and gas interfaces as critical factors in bacterial biofilm formation on magnesium implants in an animal model. Colloids Surf B Biointerfaces 2019; 186:110684. [PMID: 31812076 DOI: 10.1016/j.colsurfb.2019.110684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/09/2019] [Accepted: 11/27/2019] [Indexed: 01/23/2023]
Abstract
Infections of medical implants caused by bacterial biofilms are a major clinical problem. Bacterial colonization is predicted to be prevented by alkaline magnesium surfaces. However, in experimental animal studies, magnesium implants prolonged infections. The reason for this peculiarity likely lies within the ‒still largely hypothetical‒ mechanism by which infection arises. Investigating subcutaneous magnesium implants infected with bioluminescent Pseudomonas aeruginosa via in vivo imaging, we found that the rate of implant infections was critically dependent on a surprisingly high quantity of injected bacteria. At high inocula, bacteria were antibiotic-refractory immediately after infection. High cell densities are known to limit nutrient availability, restricting proliferation and trigger quorum sensing which could both contribute to the rapid initial resistance. We propose that gas bubbles such as those formed during magnesium corrosion, can then act as interfaces that support biofilm formation and permit long-term survival. This model could provide an explanation for the apparent ineffectiveness of innovative contact-dependent bactericidal implant surfaces in patients. In addition, the model points toward air bubbles in tissue, either by inclusion during surgery or by spontaneous gas bubble formation later on, could constitute a key risk factor for clinical implant infections.
Collapse
Affiliation(s)
- Muhammad Imran Rahim
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany; Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| | - Szymon P Szafrański
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany; Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Alexandra Ingendoh-Tsakmakidis
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany; Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Meike Stiesch
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany; Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Peter P Mueller
- Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
214
|
Byvalov AA, Konyshev IV. Yersinia pseudotuberculosis-derived adhesins. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2019. [DOI: 10.15789/2220-7619-2019-3-4-437-448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Around fifteen surface components referred to adhesins have been identified in Yersinia pseudotuberculosis combining primarily microbiological, molecular and genetic, as well as immunochemical and biophysical methods. Y. pseudotuberculosis-derived adhesins vary in structure and chemical composition but they are mainly presented by protein molecules. Some of them were shown to participate not only in adhesive but in other pathogen-related physiological functions in the host-parasite interplay. Adhesins can mediate bacterial adhesion to eukaryotic cell either directly or via the extracellular matrix components. These adhesion molecules are encoded by chromosomal DNA excepting YadA protein which gene is located in the calcium-dependence plasmid pYV common for pathogenic yersisniae. An optimum temperature for adhesin biosynthesis is located close to the body temperature of warm-blooded animals; however, at low temperature only invasin InvA, full-length smooth lipopolysaccharide and porin OmpF are produced in Y. pseudotuberculosis. Several adhesins (Psa, InvA) can be expressed at low pH (corresponds to intracellular content), thereby defining pathogenic yersiniae as facultative intracellular parasites. Three human Yersinia genus pathogens differ by ability to produce adhesins. Y. pseudotuberculosis adherence to host cells or extracellular matrix components is determined by a cumulative adhesion-based activity, which expression depends on chemical composition and physicochemical environmental conditions. It’s proposed that at the initial stage of infectious process adherence of Y. pseudotuberculosis to intestinal epithelium is mediated by InvA protein and “smooth” LPS form. These adhesins are produced in bacterial cells at low (lower than 30°С) temperature occurring in environment from which a pathogen invades into the host. At later stages of pathogenesis, after penetrating through intestinal epithelium, bacterial cells produce other adhesins, which promote survival and dissemination primarily into the mesenteric lymph nodes and, possibly, liver and spleen. At later stages of pathogenesis, after penetrating through intestinal epithelium, bacterial cells produce other adhesins, which promote survival and dissemination primarily into the mesenteric lymph nodes and, perhaps, liver and spleen. Qualitative and quantitative spectrum of Y. pseudotuberculosis adhesins is determined by environmental parameters (intercellular space, intracellular content within the diverse eukaryotic cells).
Collapse
|
215
|
Campoccia D, Mirzaei R, Montanaro L, Arciola CR. Hijacking of immune defences by biofilms: a multifront strategy. BIOFOULING 2019; 35:1055-1074. [PMID: 31762334 DOI: 10.1080/08927014.2019.1689964] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/05/2019] [Accepted: 11/04/2019] [Indexed: 06/10/2023]
Abstract
Biofilm formation by pathogens and opportunistic bacteria is the basis of persistent or recurrent infections. Up to 80% of bacterial infections in humans are associated with biofilms. Despite the efficiency of the evolved and complex human defence system against planktonic bacteria, biofilms are capable of subverting host defences. The immune system is not completely effective in opposing bacteria and preventing infection. Increasing attention is being focussed on the mechanisms enabling bacterial biofilms to skew the coordinate action of humoral and cell mediated responses. Knowledge of the interactions between biofilm bacteria and the immune system is critical to effectively address biofilm infections, which have multiplied over the years with the spread of biomaterials in medicine. In this article, the latest information on the interactions between bacterial biofilms and immune cells is examined and the areas where of information is still lacking are explored.
Collapse
Affiliation(s)
- Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Rasoul Mirzaei
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lucio Montanaro
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Carla Renata Arciola
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
216
|
Gholami SA, Goli HR, Haghshenas MR, Mirzaei B. Evaluation of polysaccharide intercellular adhesion (PIA) and glycerol teichoic acid (Gly-TA) arisen antibodies to prevention of biofilm formation in Staphylococcus aureus and Staphylococcus epidermidis strains. BMC Res Notes 2019; 12:691. [PMID: 31653277 PMCID: PMC6815028 DOI: 10.1186/s13104-019-4736-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/15/2019] [Indexed: 02/21/2023] Open
Abstract
Objective Staphylococcus aureus and S. epidermidis as opportunistic pathogens, notable for their frequency and severity of infections are recognized as the most usual reasons for medical device-associated infections that strike hospitalized patients and also immunocompromised individuals. In this study, the polysaccharide intercellular adhesion (PIA) and Glycerol teichoic acid) Gly-TA) as two major macromolecules in the biofilm formation process were purified under the native condition and their structure was analyzed by using colorimetric assays and Fourier Transform Infrared spectroscopy (FTIR). Afterward, the immune response of macromolecules and the mixture of them were assessed by measuring total IgG titers. Subsequently, biofilm inhibitory effects of raising antibodies to biofilm former S. aureus and S. epidermidis were evaluated. Results Obtained data were shown a significant rise in levels of antibodies in immunized mice with mentioned antibodies in comparison with the control group. According to the obtained findings, mentioned antibodies could eliminate S. aureus and S. epidermidis biofilm formation in vitro assays. This survey confirms the proposal that immunization of mice with a mixture of Gly-TA and PIA vaccine could be secure and protected against S. epidermidis and S. aureus infection.
Collapse
Affiliation(s)
- Sanaz Amir Gholami
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Hamid Reza Goli
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Mohammad Reza Haghshenas
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Bahman Mirzaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran. .,Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran.
| |
Collapse
|
217
|
Kandel CE, Jenkinson R, Daneman N, Backstein D, Hansen BE, Muller MP, Katz KC, Widdifield J, Bogoch E, Ward S, Sajja A, Jeldes FG, McGeer A. Predictors of Treatment Failure for Hip and Knee Prosthetic Joint Infections in the Setting of 1- and 2-Stage Exchange Arthroplasty: A Multicenter Retrospective Cohort. Open Forum Infect Dis 2019; 6:ofz452. [PMID: 31737739 PMCID: PMC6847009 DOI: 10.1093/ofid/ofz452] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/16/2019] [Indexed: 11/18/2022] Open
Abstract
Background Prosthetic hip and knee joint infections (PJIs) are challenging to eradicate despite prosthesis removal and antibiotic therapy. There is a need to understand risk factors for PJI treatment failure in the setting of prosthesis removal. Methods A retrospective cohort of individuals who underwent prosthesis removal for a PJI at 5 hospitals in Toronto, Canada, from 2010 to 2014 was created. Treatment failure was defined as recurrent PJI, amputation, death, or chronic antibiotic suppression. Potential risk factors for treatment failure were abstracted by chart review and assessed using a Cox proportional hazards model. Results A total of 533 individuals with prosthesis removal were followed for a median (interquartile range) of 814 (235–1530) days. A 1-stage exchange was performed in 19% (103/533), whereas a 2-stage procedure was completed in 88% (377/430). Treatment failure occurred in 24.8% (132/533) at 2 years; 53% (56/105) of recurrent PJIs were caused by a different bacterial species. At 4 years, treatment failure occurred in 36% of 1-stage and 32% of 2-stage procedures (P = .06). Characteristics associated with treatment failure included liver disease (adjusted hazard ratio [aHR], 3.12; 95% confidence interval [CI], 2.09–4.66), the presence of a sinus tract (aHR, 1.53; 95% CI, 1.12–2.10), preceding debridement with prosthesis retention (aHR, 1.68; 95% CI, 1.13–2.51), a 1-stage procedure (aHR, 1.72; 95% CI, 1.28–2.32), and infection due to Gram-negative bacilli (aHR, 1.35; 95% CI, 1.04–1.76). Conclusions Failure of PJI therapy is common, and risk factors are not easily modified. Improvements in treatment paradigms are needed, along with efforts to reduce orthopedic surgical site infections.
Collapse
Affiliation(s)
- Christopher E Kandel
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Richard Jenkinson
- Division of Orthopaedic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Division of Orthopaedic Surgery, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Nick Daneman
- Division of Infectious Diseases, Sunnybrook Health Sciences Centre, Toronto, Ontario Canada
| | - David Backstein
- Division of Orthopaedics , Sinai Health System, Toronto, Ontario, Canada.,Musculoskeletal Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - Bettina E Hansen
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada.,Division of Gastroenterology, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Matthew P Muller
- Unity Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Kevin C Katz
- North York General Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Widdifield
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Holland Bone & Joint Program, Toronto, Ontario, Canada.,ICES, Toronto, Ontario, Canada
| | - Earl Bogoch
- Division of Orthopaedic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Unity Health Network, University of Toronto, Toronto, Ontario, Canada.,Division of Orthopedics, Department of Surgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sarah Ward
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada.,Division of Orthopedics, Department of Surgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Abhilash Sajja
- Physician Assistant Program, Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Felipe Garcia Jeldes
- Department of Microbiology, CHU de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Allison McGeer
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada.,Sinai Health System, University of Toronto, Ontario, Canada
| |
Collapse
|
218
|
Hannachi N, Habib G, Camoin-Jau L. Aspirin Effect on Staphylococcus aureus-Platelet Interactions During Infectious Endocarditis. Front Med (Lausanne) 2019; 6:217. [PMID: 31681776 PMCID: PMC6803506 DOI: 10.3389/fmed.2019.00217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022] Open
Abstract
Infectious endocarditis (IE) is a rare disease associated with high mortality and morbidity rate. The platelet-bacterial interaction presents the cornerstone of the development of endocardial vegetation. The epidemiology of IE has undergone profound changes between the last and the new decade, with Staphylococcus aureus becoming the main incriminated species. Despite improvements in antibiotic and surgical therapies, embolic disorders remain highly associated with IE that can be fatal. Antiplatelet drugs have been widely proposed to overcome embolic events associated with IE. This proposal has been supported by numerous in vitro, experimental, and clinical studies. However, other studies have yielded conflicting results. In this review, we focus on the effect of aspirin on the genesis of S. aureus endocarditic vegetation, as well as on the management of embolic and hemorrhagic events related to it, starting by its influence on the platelet-bacteria interaction.
Collapse
Affiliation(s)
- Nadji Hannachi
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Gilbert Habib
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
- Département de Cardiologie, Hôpital de la Timone, AP-HM, Marseille, France
| | - Laurence Camoin-Jau
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
- Laboratoire d'Hématologie, Hôpital de la Timone, APHM, Marseille, France
| |
Collapse
|
219
|
El-Hennawy AS, Frolova E, Romney WA. Sodium bicarbonate catheter lock solution reduces hemodialysis catheter loss due to catheter-related thrombosis and blood stream infection: an open-label clinical trial. Nephrol Dial Transplant 2019; 34:1739-1745. [PMID: 30668833 PMCID: PMC6775472 DOI: 10.1093/ndt/gfy388] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is no ideal lock solution that prevents hemodialysis (HD) catheter loss due to catheter-related thrombosis (CRT) and catheter-related bloodstream infection (CRBSI). Catheter loss is associated with increased hospitalization and high inpatient costs. Sodium bicarbonate (NaHCO3) demonstrates anti-infective and anticoagulation properties with a good safety profile, making it an ideal lock solution development target.The objective of this study was to determine the safety and efficacy of using sodium bicarbonate catheter lock solution (SBCLS) as a means of preventing HD catheter loss due to CRT and CRBSI. METHODS The study took place in a community hospital in Brooklyn, NY, USA. All admitted patients ≥18 years of age who needed HD treatment through CVC were included in the study. 451 patients included in the study were provided SBCLS or NSCLS post-dialysis. Catheter loss due to CRT or CRBSI was evaluated over a period of 546 days. RESULTS A total of 452 patients met the criteria; 1 outlier was excluded, 226 were in the NSCLS group and 225 were in the SBCLS group. There were no significant differences between groups in comorbidities at the outset. The NSCLS group had CRT and CRBSI rates of 4.1 and 2.6/1000 catheter days (CD), respectively, compared with 0.17/1000 CD for both outcomes in the SBCLS group. SBCLS patients had a significantly reduced catheter loss rate due to CRT (P < 0.0001) and CRBSI (P = 0.0004). NSCLS patients had higher odds of losing their catheter due to CRT {odds ratio [OR] 26.6 [95% confidence interval (CI) 3.57-198.52]} and CRBSI [OR 15.9 (95% CI 2.09-121.61)] during the study period. CONCLUSION The novel approach of using SBCLS was found to be safe and was statistically superior to normal saline in preventing HD catheter loss due to CRT and CRBSI. NaHCO3 solution is inexpensive, readily available in various settings and holds the potential to decrease hospitalization, length of stay and dialysis-related costs. TRIAL REGISTRATION Maimonides Medical Center Investigational Review Board, Study IRB 2015-06-25-CIH. ClinicalTrials.gov identifier: NCT03627884.
Collapse
Affiliation(s)
- Adel S El-Hennawy
- Department of Nephrology, NYC Health + Hospitals/Coney Island, Brooklyn, NY, USA
| | - Elena Frolova
- Department of Nephrology, NYC Health + Hospitals/Coney Island, Brooklyn, NY, USA
| | - Wesley A Romney
- Department of Medicine, NYC Health + Hospitals/Coney Island, Brooklyn, NY, USA
| |
Collapse
|
220
|
Bottagisio M, Soggiu A, Piras C, Bidossi A, Greco V, Pieroni L, Bonizzi L, Roncada P, Lovati AB. Proteomic Analysis Reveals a Biofilm-Like Behavior of Planktonic Aggregates of Staphylococcus epidermidis Grown Under Environmental Pressure/Stress. Front Microbiol 2019; 10:1909. [PMID: 31551940 PMCID: PMC6743020 DOI: 10.3389/fmicb.2019.01909] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 08/05/2019] [Indexed: 01/14/2023] Open
Abstract
Prosthetic joint replacement failure has a huge impact on quality of life and hospitalization costs. A leading cause of prosthetic joint infection is bacteria-forming biofilm on the surface of orthopedic devices. Staphylococcus epidermidis is an emergent, low-virulence pathogen implicated in chronic infections, barely indistinguishable from aseptic loosening when embedded in a mature matrix. The literature on the behavior of quiescent S. epidermidis in mature biofilms is scarce. To fill this gap, we performed comparative analysis of the whole proteomic profiles of two methicillin-resistant S. epidermidis strains growing in planktonic and in sessile form to investigate the molecular mechanisms underlying biofilm stability. After 72-h culture of biofilm-forming S. epidermidis, overexpression of proteins involved in the synthesis of nucleoside triphosphate and polysaccharides was observed, whereas planktonic bacteria expressed proteins linked to stress and anaerobic growth. Cytological analysis was performed to determine why planktonic bacteria unexpectedly expressed proteins typical of sessile culture. Images evidenced that prolonged culture under vigorous agitation can create a stressful growing environment that triggers microorganism aggregation in a biofilm-like matrix as a mechanism to survive harsh conditions. The choice of a unique late time point provided an important clue for future investigations into the biofilm-like behavior of planktonic cells. Our preliminary results may inform comparative proteomic strategies in the study of mature bacterial biofilm. Finally, there is an increasing number of studies on the aggregation of free-floating bacteria embedded in an extracellular matrix, prompting the need to gain further insight into this mode of bacterial growth.
Collapse
Affiliation(s)
- Marta Bottagisio
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Clinical Chemistry and Microbiology, Milan, Italy
| | - Alessio Soggiu
- Department of Veterinary Medicine (DiMeVet), University of Milan, Milan, Italy
| | - Cristian Piras
- Department of Veterinary Medicine (DiMeVet), University of Milan, Milan, Italy
| | - Alessandro Bidossi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Clinical Chemistry and Microbiology, Milan, Italy
| | - Viviana Greco
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore Roma, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luisa Pieroni
- Proteomics and Metabonomics Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Luigi Bonizzi
- Department of Veterinary Medicine (DiMeVet), University of Milan, Milan, Italy
| | - Paola Roncada
- Department of Health Sciences, Università degli Studi "Magna Græcia", Catanzaro, Italy
| | - Arianna B Lovati
- IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, Milan, Italy
| |
Collapse
|
221
|
Arciola CR, Campoccia D, Montanaro L. Implant infections: adhesion, biofilm formation and immune evasion. Nat Rev Microbiol 2019; 16:397-409. [PMID: 29720707 DOI: 10.1038/s41579-018-0019-y] [Citation(s) in RCA: 1123] [Impact Index Per Article: 187.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Medical device-associated infections account for a large proportion of hospital-acquired infections. A variety of opportunistic pathogens can cause implant infections, depending on the type of the implant and on the anatomical site of implantation. The success of these versatile pathogens depends on rapid adhesion to virtually all biomaterial surfaces and survival in the hostile host environment. Biofilm formation on implant surfaces shelters the bacteria and encourages persistence of infection. Furthermore, implant-infecting bacteria can elude innate and adaptive host defences as well as biocides and antibiotic chemotherapies. In this Review, we explore the fundamental pathogenic mechanisms underlying implant infections, highlighting orthopaedic implants and Staphylococcus aureus as a prime example, and discuss innovative targets for preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy. .,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Davide Campoccia
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Lucio Montanaro
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
222
|
Di Domenico EG, Cavallo I, Capitanio B, Ascenzioni F, Pimpinelli F, Morrone A, Ensoli F. Staphylococcus aureus and the Cutaneous Microbiota Biofilms in the Pathogenesis of Atopic Dermatitis. Microorganisms 2019; 7:E301. [PMID: 31470558 PMCID: PMC6780378 DOI: 10.3390/microorganisms7090301] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/12/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022] Open
Abstract
Biofilm is the dominant mode of growth of the skin microbiota, which promotes adhesion and persistence in the cutaneous microenvironment, thus contributing to the epidermal barrier function and local immune modulation. In turn, the local immune microenvironment plays a part in shaping the skin microbiota composition. Atopic dermatitis (AD) is an immune disorder characterized by a marked dysbiosis, with a sharp decline of microbial diversity. During AD flares biofilm-growing Staphylococcus aureus emerges as the major colonizer in the skin lesions, in strict association with disease severity. The chronic production of inflammatory cytokines in the skin of AD individuals concurs at supporting S. aureus biofilm overgrowth at the expense of other microbial commensals, subverting the composition of the healthy skin microbiome. The close relationship between the host and microbial biofilm resident in the skin has profound implications on human health, making skin microbiota an attractive target for the therapeutic management of different skin disorders.
Collapse
Affiliation(s)
- Enea Gino Di Domenico
- Clinical Pathology and Microbiology, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy.
| | - Ilaria Cavallo
- Clinical Pathology and Microbiology, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy
| | - Bruno Capitanio
- Division of Dermatology, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology C. Darwin, University of Rome Sapienza, 00161 Rome, Italy
| | - Fulvia Pimpinelli
- Clinical Pathology and Microbiology, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy
| | - Aldo Morrone
- Scientific Director San Gallicano Dermatological Institute IRCCS, 00144 Rome, Italy
| | - Fabrizio Ensoli
- Clinical Pathology and Microbiology, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy
| |
Collapse
|
223
|
Gilbertie JM, Schnabel LV, Hickok NJ, Jacob ME, Conlon BP, Shapiro IM, Parvizi J, Schaer TP. Equine or porcine synovial fluid as a novel ex vivo model for the study of bacterial free-floating biofilms that form in human joint infections. PLoS One 2019; 14:e0221012. [PMID: 31415623 PMCID: PMC6695105 DOI: 10.1371/journal.pone.0221012] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
Bacterial invasion of synovial joints, as in infectious or septic arthritis, can be difficult to treat in both veterinary and human clinical practice. Biofilms, in the form of free-floating clumps or aggregates, are involved with the pathogenesis of infectious arthritis and periprosthetic joint infection (PJI). Infection of a joint containing an orthopedic implant can additionally complicate these infections due to the presence of adherent biofilms. Because of these biofilm phenotypes, bacteria within these infected joints show increased antimicrobial tolerance even at high antibiotic concentrations. To date, animal models of PJI or infectious arthritis have been limited to small animals such as rodents or rabbits. Small animal models, however, yield limited quantities of synovial fluid making them impractical for in vitro research. Herein, we describe the use of ex vivo equine and porcine models for the study of synovial fluid induced biofilm aggregate formation and antimicrobial tolerance. We observed Staphylococcus aureus and other bacterial pathogens adapt the same biofilm aggregate phenotype with significant antimicrobial tolerance in both equine and porcine synovial fluid, analogous to human synovial fluid. We also demonstrate that enzymatic dispersal of synovial fluid aggregates restores the activity of antimicrobials. Future studies investigating the interaction of bacterial cell surface proteins with host synovial fluid proteins can be readily carried out in equine or porcine ex vivo models to identify novel drug targets for treatment of prevention of these difficult to treat infectious diseases.
Collapse
Affiliation(s)
- Jessica M. Gilbertie
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
- Department of Clinical Studies New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, United States of America
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
| | - Noreen J. Hickok
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Megan E. Jacob
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, United States of America
| | - Irving M. Shapiro
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Javad Parvizi
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Thomas P. Schaer
- Department of Clinical Studies New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, United States of America
| |
Collapse
|
224
|
Cutrona N, Gillard K, Ulrich R, Seemann M, Miller HB, Blackledge MS. From Antihistamine to Anti-infective: Loratadine Inhibition of Regulatory PASTA Kinases in Staphylococci Reduces Biofilm Formation and Potentiates β-Lactam Antibiotics and Vancomycin in Resistant Strains of Staphylococcus aureus. ACS Infect Dis 2019; 5:1397-1410. [PMID: 31132246 DOI: 10.1021/acsinfecdis.9b00096] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Staphylococcus epidermidis and Staphylococcus aureus are important human pathogens responsible for two-thirds of all postsurgical infections of indwelling medical devices. Staphylococci form robust biofilms that provide a reservoir for chronic infection, and antibiotic-resistant isolates are increasingly common in both healthcare and community settings. Novel treatments that can simultaneously inhibit biofilm formation and antibiotic-resistance pathways are urgently needed to combat the increasing rates of antibiotic-resistant infections. Herein we report that loratadine, an FDA-approved antihistamine, significantly inhibits biofilm formation in both S. aureus and S. epidermidis. Furthermore, loratadine potentiates β-lactam antibiotics in methicillin-resistant strains of S. aureus and potentiates both β-lactam antibiotics and vancomycin in vancomycin-resistant strains of S. aureus. Additionally, we elucidate loratadine's mechanism of action as a novel inhibitor of the regulatory PASTA kinases Stk and Stk1 in S. epidermidis and S. aureus, respectively. Finally, we describe how Stk1 inhibition affects the expression of genes involved in both biofilm formation and antibiotic resistance in S. epidermidis and S. aureus.
Collapse
Affiliation(s)
- Nicholas Cutrona
- Department of Chemistry, High Point University, One University Parkway, High Point, North Carolina 27268, United States
| | - Kyra Gillard
- Department of Chemistry, High Point University, One University Parkway, High Point, North Carolina 27268, United States
| | - Rebecca Ulrich
- Department of Chemistry, High Point University, One University Parkway, High Point, North Carolina 27268, United States
| | - Mikaela Seemann
- Department of Chemistry, High Point University, One University Parkway, High Point, North Carolina 27268, United States
| | - Heather B. Miller
- Department of Chemistry, High Point University, One University Parkway, High Point, North Carolina 27268, United States
| | - Meghan S. Blackledge
- Department of Chemistry, High Point University, One University Parkway, High Point, North Carolina 27268, United States
| |
Collapse
|
225
|
Peng C, Hanawa T, Azam AH, LeBlanc C, Ung P, Matsuda T, Onishi H, Miyanaga K, Tanji Y. Silviavirus phage ɸMR003 displays a broad host range against methicillin-resistant Staphylococcus aureus of human origin. Appl Microbiol Biotechnol 2019; 103:7751-7765. [PMID: 31388727 DOI: 10.1007/s00253-019-10039-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/05/2019] [Accepted: 07/14/2019] [Indexed: 12/17/2022]
Abstract
The emergence of life-threatening methicillin-resistant Staphylococcus aureus (MRSA) has led to increased interest in the use of bacteriophages as an alternative therapy to antibiotics. The success of phage therapy is greatly dependent on the selected phage possessing a wide host range. This study describes phage ɸMR003 isolated from sewage influent at a municipal wastewater treatment plant in Tokyo, Japan. ɸMR003 could infect 97% of 104 healthcare- and community-associated MRSA strains tested, compared with 73% for phage ɸSA012, which has a broad host range against bovine mastitis S. aureus. Genome analysis revealed that ɸMR003 belongs to the genus Silviavirus which has not been studied extensively. ɸMR003 recognizes and binds to wall teichoic acid (WTA) of S. aureus during infection. In silico comparisons of the genomes of ɸMR003 and ɸSA012 revealed that ORF117 and ORF119 of ɸMR003 are homologous to the putative receptor-binding proteins ORF103 and ORF105 of ɸSA012, with amino acid similarities of 75% and 72%, respectively. ORF104, which is an N-acetylglucosaminidase found in the ɸMR003 tail, may facilitate phage's infection onto the WTA-null S. aureus RN4220. The differences in tail and baseplate proteins may be key contributing factors to the different host specificities of ɸMR003 and ɸSA012. ɸMR003 showed strong adsorptivity, but not infectivity, against S. aureus SA003, which may be influenced by the bacterium's restriction modification system. This study expands our knowledge of the genomic diversity and host specificity of Silviavirus, which is a potential phage therapy candidate for MRSA infections.
Collapse
Affiliation(s)
- Chanthol Peng
- School of Life Science and Technology, Tokyo Institute of Technology, 4259J2-15 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
- Faculty of Chemical and Food Engineering, Institute of Technology of Cambodia, Russian Federation Blvd., P.O. Box 86, Phnom Penh, 12156, Cambodia
| | - Tomoko Hanawa
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Aa Haeruman Azam
- School of Life Science and Technology, Tokyo Institute of Technology, 4259J2-15 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Cierra LeBlanc
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Porsry Ung
- Faculty of Chemical and Food Engineering, Institute of Technology of Cambodia, Russian Federation Blvd., P.O. Box 86, Phnom Penh, 12156, Cambodia
| | - Takeaki Matsuda
- Department of Traumatology and Critical Care Medicine, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Hiroaki Onishi
- Department of Laboratory Medicine, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, 4259J2-15 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Yasunori Tanji
- School of Life Science and Technology, Tokyo Institute of Technology, 4259J2-15 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.
| |
Collapse
|
226
|
Josse J, Valour F, Maali Y, Diot A, Batailler C, Ferry T, Laurent F. Interaction Between Staphylococcal Biofilm and Bone: How Does the Presence of Biofilm Promote Prosthesis Loosening? Front Microbiol 2019; 10:1602. [PMID: 31379772 PMCID: PMC6653651 DOI: 10.3389/fmicb.2019.01602] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/26/2019] [Indexed: 12/19/2022] Open
Abstract
With the aging of population, the number of indications for total joint replacement is continuously increasing. However, prosthesis loosening can happen and is related to two major mechanisms: (1) aseptic loosening due to prosthesis micromotion and/or corrosion and release of wear particles from the different components of the implanted material and (2) septic loosening due to chronic prosthetic joint infection (PJI). The “aseptic” character of prosthesis loosening has been challenged over the years, especially considering that bacteria can persist in biofilms and be overlooked during diagnosis. Histological studies on periprosthetic tissue samples reported that macrophages are the principle cells associated with aseptic loosening due to wear debris. They produce cytokines and favor an inflammatory environment that induces formation and activation of osteoclasts, leading to bone resorption and periprosthetic osteolysis. In PJIs, the presence of infiltrates of polymorphonuclear neutrophils is a major criterion for histological diagnosis. Neutrophils are colocalized with osteoclasts and zones of osteolysis. A similar inflammatory environment also develops, leading to bone resorption through osteoclasts. Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis are the main staphylococci observed in PJIs. They share the common feature to form biofilm. For S. aureus and S. epidermidis, the interaction between biofilm and immunes cells (macrophages and polymorphonuclear neutrophils) differs regarding the species. Indeed, the composition of extracellular matrix of biofilm seems to impact the interaction with immune cells. Recent papers also reported the major role of myeloid-derived suppressor cells in biofilm-associated PJIs with S. aureus. These cells prevent lymphocyte infiltration and facilitate biofilm persistence. Moreover, the role of T lymphocytes is still unclear and potentially underestimates. In this review, after introducing the cellular mechanism of aseptic and septic loosening, we will focus on the interrelationships between staphylococcal biofilm, immune cells, and bone cells.
Collapse
Affiliation(s)
- Jérôme Josse
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Yousef Maali
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Alan Diot
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Cécile Batailler
- Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Maladies Infectieuses, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Laboratoire de Bactériologie, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
227
|
Voorhies AA, Mark Ott C, Mehta S, Pierson DL, Crucian BE, Feiveson A, Oubre CM, Torralba M, Moncera K, Zhang Y, Zurek E, Lorenzi HA. Study of the impact of long-duration space missions at the International Space Station on the astronaut microbiome. Sci Rep 2019; 9:9911. [PMID: 31289321 PMCID: PMC6616552 DOI: 10.1038/s41598-019-46303-8] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/17/2019] [Indexed: 12/16/2022] Open
Abstract
Over the course of a mission to the International Space Station (ISS) crew members are exposed to a number of stressors that can potentially alter the composition of their microbiomes and may have a negative impact on astronauts’ health. Here we investigated the impact of long-term space exploration on the microbiome of nine astronauts that spent six to twelve months in the ISS. We present evidence showing that the microbial communities of the gastrointestinal tract, skin, nose and tongue change during the space mission. The composition of the intestinal microbiota became more similar across astronauts in space, mostly due to a drop in the abundance of a few bacterial taxa, some of which were also correlated with changes in the cytokine profile of crewmembers. Alterations in the skin microbiome that might contribute to the high frequency of skin rashes/hypersensitivity episodes experienced by astronauts in space were also observed. The results from this study demonstrate that the composition of the astronauts’ microbiome is altered during space travel. The impact of those changes on crew health warrants further investigation before humans embark on long-duration voyages into outer space.
Collapse
Affiliation(s)
- Alexander A Voorhies
- Department of Infectious Diseases, J. Craig Venter Institute, Rockville, MD, USA
| | - C Mark Ott
- NASA-Johnson Space Center, Houston, TX, USA
| | | | | | | | | | | | - Manolito Torralba
- Department of Infectious Diseases, J. Craig Venter Institute, Rockville, MD, USA
| | - Kelvin Moncera
- Department of Infectious Diseases, J. Craig Venter Institute, Rockville, MD, USA
| | - Yun Zhang
- Department of Infectious Diseases, J. Craig Venter Institute, Rockville, MD, USA
| | | | - Hernan A Lorenzi
- Department of Infectious Diseases, J. Craig Venter Institute, Rockville, MD, USA.
| |
Collapse
|
228
|
Bahonar S, Ghazvinian M, Haghshenas MR, Goli HR, Mirzaei B. Purification of PIA and rSesC as Putative Vaccine Candidates Against Staphylococcus aureus. Rep Biochem Mol Biol 2019; 8:161-167. [PMID: 31832440 PMCID: PMC6844615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Staphylococcus aureus is predominant at sites of biomaterial-associated infection (BAI) and frequently infects hospitalized individuals. METHODS The polysaccharide intercellular adhesin (PIA) and S. epidermidis rSesC protein, major macromolecules in biofilm formation, were purified under native conditions and cloned and expressed in a prokaryotic host. RESULTS LPurification of the macromolecules was confirmed by FTIR and Western blotting. CONCLUSION The S. epidermidis SesC protein and PIA were uccessfully purified. Both are considered as vaccine candidates.
Collapse
Affiliation(s)
- Sara Bahonar
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
- Molecular and cell biology Research center, Faculty of Medicine, Mazandaran University of Medical sciences, Sari, Iran.
| | - Maryam Ghazvinian
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
| | - Mohamad Reza Haghshenas
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
| | - Hamid Reza Goli
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
| | - Bahman Mirzaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
- Molecular and cell biology Research center, Faculty of Medicine, Mazandaran University of Medical sciences, Sari, Iran.
| |
Collapse
|
229
|
Graf AC, Leonard A, Schäuble M, Rieckmann LM, Hoyer J, Maass S, Lalk M, Becher D, Pané-Farré J, Riedel K. Virulence Factors Produced by Staphylococcus aureus Biofilms Have a Moonlighting Function Contributing to Biofilm Integrity. Mol Cell Proteomics 2019; 18:1036-1053. [PMID: 30850421 PMCID: PMC6553939 DOI: 10.1074/mcp.ra118.001120] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/19/2019] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus is the causative agent of various biofilm-associated infections in humans causing major healthcare problems worldwide. This type of infection is inherently difficult to treat because of a reduced metabolic activity of biofilm-embedded cells and the protective nature of a surrounding extracellular matrix (ECM). However, little is known about S. aureus biofilm physiology and the proteinaceous composition of the ECM. Thus, we cultivated S. aureus biofilms in a flow system and comprehensively profiled intracellular and extracellular (ECM and flow-through (FT)) biofilm proteomes, as well as the extracellular metabolome compared with planktonic cultures. Our analyses revealed the expression of many pathogenicity factors within S. aureus biofilms as indicated by a high abundance of capsule biosynthesis proteins along with various secreted virulence factors, including hemolysins, leukotoxins, and lipases as a part of the ECM. The activity of ECM virulence factors was confirmed in a hemolysis assay and a Galleria mellonella pathogenicity model. In addition, we uncovered a so far unacknowledged moonlighting function of secreted virulence factors and ribosomal proteins trapped in the ECM: namely their contribution to biofilm integrity. Mechanistically, it was revealed that this stabilizing effect is mediated by the strong positive charge of alkaline virulence factors and ribosomal proteins in an acidic ECM environment, which is caused by the release of fermentation products like formate, lactate, and acetate because of oxygen limitation in biofilms. The strong positive charge of these proteins most likely mediates electrostatic interactions with anionic cell surface components, eDNA, and anionic metabolites. In consequence, this leads to strong cell aggregation and biofilm stabilization. Collectively, our study identified a new molecular mechanism during S. aureus biofilm formation and thus significantly widens the understanding of biofilm-associated S. aureus infections - an essential prerequisite for the development of novel antimicrobial therapies.
Collapse
Affiliation(s)
- Alexander C Graf
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Anne Leonard
- §Institute of Biochemistry, Department of Cellular Biochemistry and Metabolomics
| | - Manuel Schäuble
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Lisa M Rieckmann
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Juliane Hoyer
- ¶Institute of Microbiology, Department of Microbial Proteomics; University of Greifswald, Germany
| | - Sandra Maass
- ¶Institute of Microbiology, Department of Microbial Proteomics; University of Greifswald, Germany
| | - Michael Lalk
- §Institute of Biochemistry, Department of Cellular Biochemistry and Metabolomics
| | - Dörte Becher
- ¶Institute of Microbiology, Department of Microbial Proteomics; University of Greifswald, Germany
| | - Jan Pané-Farré
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology
| | - Katharina Riedel
- From the ‡Institute of Microbiology, Department of Microbial Physiology and Molecular Biology;
| |
Collapse
|
230
|
Souza dos Santos B, Bezerra Filho CM, Alves do Nascimento Junior JA, Brust FR, Bezerra-Silva PC, Lino da Rocha SK, Krogfelt KA, Maria do Amaral Ferraz Navarro D, Tereza dos Santos Correia M, Napoleão TH, Nascimento da Silva LC, Macedo AJ, Vanusa da Silva M, Guedes Paiva PM. Anti-staphylococcal activity of Syagrus coronata essential oil: Biofilm eradication and in vivo action on Galleria mellonela infection model. Microb Pathog 2019; 131:150-157. [DOI: 10.1016/j.micpath.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 01/31/2023]
|
231
|
Shanmugam N, Baker MODG, Ball SR, Steain M, Pham CLL, Sunde M. Microbial functional amyloids serve diverse purposes for structure, adhesion and defence. Biophys Rev 2019; 11:287-302. [PMID: 31049855 PMCID: PMC6557962 DOI: 10.1007/s12551-019-00526-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The functional amyloid state of proteins has in recent years garnered much attention for its role in serving crucial and diverse biological roles. Amyloid is a protein fold characterised by fibrillar morphology, binding of the amyloid-specific dyes Thioflavin T and Congo Red, insolubility and underlying cross-β structure. Amyloids were initially characterised as an aberrant protein fold associated with mammalian disease. However, in the last two decades, functional amyloids have been described in almost all biological systems, from viruses, to bacteria and archaea, to humans. Understanding the structure and role of these amyloids elucidates novel and potentially ancient mechanisms of protein function throughout nature. Many of these microbial functional amyloids are utilised by pathogens for invasion and maintenance of infection. As such, they offer novel avenues for therapies. This review examines the structure and mechanism of known microbial functional amyloids, with a particular focus on the pathogenicity conferred by the production of these structures and the strategies utilised by microbes to interfere with host amyloid structures. The biological importance of microbial amyloid assemblies is highlighted by their ubiquity and diverse functionality.
Collapse
Affiliation(s)
- Nirukshan Shanmugam
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health and Sydney Nano, University of Sydney, Sydney, NSW, 2006, Australia
| | - Max O D G Baker
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health and Sydney Nano, University of Sydney, Sydney, NSW, 2006, Australia
| | - Sarah R Ball
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health and Sydney Nano, University of Sydney, Sydney, NSW, 2006, Australia
| | - Megan Steain
- Infectious Diseases and Immunology, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Chi L L Pham
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health and Sydney Nano, University of Sydney, Sydney, NSW, 2006, Australia
| | - Margaret Sunde
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health and Sydney Nano, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
232
|
Postantibiotic and Sub-MIC Effects of Exebacase (Lysin CF-301) Enhance Antimicrobial Activity against Staphylococcus aureus. Antimicrob Agents Chemother 2019; 63:AAC.02616-18. [PMID: 30936103 DOI: 10.1128/aac.02616-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/24/2019] [Indexed: 12/13/2022] Open
Abstract
CF-301 (exebacase) is a recombinantly produced bacteriophage-derived lysin (cell wall hydrolase) and is the first agent of this class to enter clinical development in the United States for treating bacteremia including endocarditis due to Staphylococcus aureus Whereas rapid bactericidal activity is the hallmark in vitro and in vivo response to CF-301 at exposures higher than the MIC, prolonged antimicrobial activity, mediated by cell wall damage, is predicted at concentrations less than the MIC. In the current study, a series of in vitro pharmacodynamic parameters, including the postantibiotic effect (PAE), postantibiotic sub-MIC effect (PA-SME), and sub-MIC effect (SME), were studied to determine how short-duration and sub-MIC CF-301 exposures affect the growth of surviving staphylococci and extend its antimicrobial activity. Mean PAE, PA-SME, and SME values up to 4.8, 9.3, and 9.8 h, respectively, were observed against 14 staphylococcal strains tested in human serum; growth delays were extended by 6 h in the presence of daptomycin. Exposures to CF-301 at sub-MIC levels as low as 0.001× to 0.01× MIC (∼1 to 10 ng/ml) resulted in aberrant cell wall ultrastructure, increased membrane permeability, dissipation of membrane potential, and inhibition of virulence phenotypes, including agglutination and biofilm formation. A mouse thigh infection model designed to study the PAE was used to confirm our findings and demonstrate in vivo growth delays of ≥19.3 h. Our findings suggest that at CF-301 concentrations less than the MIC during therapeutic use, sustained reductions in bacterial fitness and virulence may substantially enhance efficacy.
Collapse
|
233
|
Kwiecinski JM, Crosby HA, Valotteau C, Hippensteel JA, Nayak MK, Chauhan AK, Schmidt EP, Dufrêne YF, Horswill AR. Staphylococcus aureus adhesion in endovascular infections is controlled by the ArlRS-MgrA signaling cascade. PLoS Pathog 2019; 15:e1007800. [PMID: 31116795 PMCID: PMC6548404 DOI: 10.1371/journal.ppat.1007800] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/04/2019] [Accepted: 04/30/2019] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is a leading cause of endovascular infections. This bacterial pathogen uses a diverse array of surface adhesins to clump in blood and adhere to vessel walls, leading to endothelial damage, development of intravascular vegetations and secondary infectious foci, and overall disease progression. In this work, we describe a novel strategy used by S. aureus to control adhesion and clumping through activity of the ArlRS two-component regulatory system, and its downstream effector MgrA. Utilizing a combination of in vitro cellular assays, and single-cell atomic force microscopy, we demonstrated that inactivation of this ArlRS—MgrA cascade inhibits S. aureus adhesion to a vast array of relevant host molecules (fibrinogen, fibronectin, von Willebrand factor, collagen), its clumping with fibrinogen, and its attachment to human endothelial cells and vascular structures. This impact on S. aureus adhesion was apparent in low shear environments, and in physiological levels of shear stress, as well as in vivo in mouse models. These effects were likely mediated by the de-repression of giant surface proteins Ebh, SraP, and SasG, caused by inactivation of the ArlRS—MgrA cascade. In our in vitro assays, these giant proteins collectively shielded the function of other surface adhesins and impaired their binding to cognate ligands. Finally, we demonstrated that the ArlRS—MgrA regulatory cascade is a druggable target through the identification of a small-molecule inhibitor of ArlRS signaling. Our findings suggest a novel approach for the pharmacological treatment and prevention of S. aureus endovascular infections through targeting the ArlRS—MgrA regulatory system. Adhesion is central to the success of Staphylococcus aureus as a bacterial pathogen. We describe a novel mechanism through which S. aureus alters adhesion to ligands by regulating expression of giant inhibitory surface proteins. These giant proteins shield normal surface adhesins, preventing binding to ligands commonly found in the bloodstream and vessel walls. Using this unique regulatory scheme, S. aureus can bypass the need for individualized regulation of numerous adhesins to control overall adhesive properties. Our study establishes the importance of these giant proteins for S. aureus pathogenesis and demonstrates that a single regulatory cascade can be targeted for treating infections.
Collapse
Affiliation(s)
- Jakub M. Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Heidi A. Crosby
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Claire Valotteau
- Institute of Life Sciences, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Joseph A. Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Manasa K. Nayak
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Anil K. Chauhan
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Eric P. Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Yves F. Dufrêne
- Institute of Life Sciences, Université catholique de Louvain, Louvain-la-Neuve, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
234
|
Gutierrez Jauregui R, Fleige H, Bubke A, Rohde M, Weiss S, Förster R. IL-1β Promotes Staphylococcus aureus Biofilms on Implants in vivo. Front Immunol 2019; 10:1082. [PMID: 31156635 PMCID: PMC6534041 DOI: 10.3389/fimmu.2019.01082] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022] Open
Abstract
Implant associated infections represent a serious health burden in clinics since some microorganisms are able to colonize biological surfaces or surfaces of indwelling medical devices and form biofilms. Biofilms represent communities of microorganisms attached to hydrated surfaces and enclosed in self-produced extracellular matrix. This renders them resistant to exogenous assaults like antibiotics or immune effector mechanisms. Little is known regarding the role of the immune system in the formation of biofilms during implant associated infections, largely due to the lack of suitable mouse models. Here we use colonized osmotic pumps in mice to study the interaction of an activated immune system with biofilm-forming Staphylococcus aureus encoding Gaussia luciferase. This approach permits biofilm formation on the osmotic pumps in living animals. It also allows the continuous supply of soluble immune cell activating agents, such as cytokines to study their effect on biofilm formation in vivo. Using non-invasive imaging of the bioluminescent signal emitted by the lux expressing bacteria for quantification of bacterial load in conjunction with light and electron microscopy, we observed that pump-supplied pro-inflammatory cytokine IL-1β strongly increased biofilm formation along with a massive influx of neutrophils adjacent to the biofilm-coated pumps. Thus, our data demonstrate that immune defense mechanisms can augment biofilm formation.
Collapse
Affiliation(s)
| | - Henrike Fleige
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Anja Bubke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholz Center for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,RESIST, Cluster of Excellence 2155, Hannover Medical School, Hannover, Germany
| |
Collapse
|
235
|
MroQ Is a Novel Abi-Domain Protein That Influences Virulence Gene Expression in Staphylococcus aureus via Modulation of Agr Activity. Infect Immun 2019; 87:IAI.00002-19. [PMID: 30833335 DOI: 10.1128/iai.00002-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/23/2019] [Indexed: 01/07/2023] Open
Abstract
Numerous factors have, to date, been identified as playing a role in the regulation of Agr activity in Staphylococcus aureus, including transcription factors, antisense RNAs, and host elements. Herein we investigated the product of SAUSA300_1984 (termed MroQ), a transmembrane Abi-domain/M79 protease-family protein, as a novel effector of this system. Using a USA300 mroQ mutant, we observed a drastic reduction in proteolysis, hemolysis, and pigmentation that was fully complementable. This appears to result from diminished agr activity, as transcriptional analysis revealed significant decreases in expression of both RNAII and RNAIII in the mroQ mutant. Such effects appear to be direct, rather than indirect, as known agr effectors demonstrated limited alterations in their activity upon mroQ disruption. A comparison of RNA sequencing data sets for both mroQ and agr mutants revealed a profound overlap in their regulomes, with the majority of factors affected being known virulence determinants. Importantly, the preponderance of alterations in expression were more striking in the agr mutant, indicating that MroQ is necessary, but not sufficient, for Agr function. Mechanism profiling revealed that putative residues for metalloprotease activity within MroQ are required for its Agr-controlling effect; however, this was not wielded at the level of AgrD processing. Virulence assessment demonstrated that both mroQ and agr mutants exhibited increased formation of renal abscesses but decreased skin abscess formation alongside diminished dermonecrosis. Collectively, we present the characterization of a novel agr effector in S. aureus which would appear to be a direct regulator, potentially functioning via interaction with the AgrC histidine kinase.
Collapse
|
236
|
Jasmine S, A T, Janarthanan K, Krishnamoorthy R, Alshatwi AA. Antimicrobial and antibiofilm potential of injectable platelet rich fibrin-a second-generation platelet concentrate-against biofilm producing oral staphylococcus isolates. Saudi J Biol Sci 2019; 27:41-46. [PMID: 31889815 PMCID: PMC6933211 DOI: 10.1016/j.sjbs.2019.04.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 01/15/2023] Open
Abstract
Injectable Platelet rich fibrin (i-PRF) is a platelet concentrate that has been extensively used for multiple medical purposes and is a valuable adjunct for the regeneration of damaged tissues in surgical procedures. The enriched bioactive substances in i-PRF are responsible for speeding the wound healing process. Infection of biofilm producing bacteria in surgical wounds is becoming a serious threat. Research in this field is focused on new strategies to fight infections and to reduce the healing time. The present study was aimed to evaluate the in vitro antimicrobial and antibiofilm effects of i-PRF against oral pathogenic biofilm producing staphylococcus bacteria isolated from patient with dental and oral abscess. The antibacterial activity of i-PRF, was determined through broth microdilution as minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC). i-PRF exhibited bactericidal activity against both non biofilm and biofilm producing bacteria. i-PRF could be potential antimicrobial peptide used to combat postoperative infections caused by biofilm producing staphylococcus.
Collapse
Affiliation(s)
- Sharmila Jasmine
- Department of Oral Maxillofacial Surgery, Rajah Muthiah Dental College and Hospital, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Thangavelu A
- Department of Oral Maxillofacial Surgery, Rajah Muthiah Dental College and Hospital, Annamalai University, Chidambaram, Tamil Nadu, India
| | - K Janarthanan
- Department of Oral Maxillofacial Surgery, Rajah Muthiah Dental College and Hospital, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Rajapandiyan Krishnamoorthy
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia
| | - Ali A Alshatwi
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia
| |
Collapse
|
237
|
Gharaibeh MH, Khalifeh MS, Zattout EM, Abu-Qatouseh LF. Potential antimicrobial effect of plant essential oils and virulence genes expression in methicillin-resistant Staphylococcus aureus isolates. Vet World 2019; 13:669-675. [PMID: 32546910 PMCID: PMC7245711 DOI: 10.14202/vetworld.2020.669-675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/04/2020] [Indexed: 12/24/2022] Open
Abstract
AIM This study aimed to investigate the antibacterial efficacy of eight commercially available essential oil (EO) blends and characterize the effect on the expression of some virulence genes against methicillin-resistant Staphylococcus aureus (MRSA). MATERIALS AND METHODS In vitro evaluation of the antimicrobial effects of oils against MRSA was performed using the disk diffusion method and by measuring the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC). The EOs (A-F) were contained (β-pinene, carvacrol, carvone, dimethyl trisulfide, linalool, limonene, menthol, monoterpene hydrocarbons, and thymol) in different amounts. In addition, a real-time polymerase chain reaction was also used to determine the gene expression of the virulence genes (intercellular adhesion cluster [ica]-9, ica-15, and RNA III) against MRSA (ATCC 43300) after treatment with selected oils. RESULTS Among the eight EOs evaluated, EO (D), (E), and (A) showed, in general, the greatest antimicrobial activity against MRSA. EO at 1/3 MIC has effectively down-regulated ica-9 and ica-15 of MRSA by 17.83 and 4.94 folds, respectively. Meanwhile, EO (A) has effectively down-regulated RNAIII by 3.74 folds. Our results indicated that some of the EOs exhibit promising antimicrobial effects against MRSA isolates. Moreover, the results of the analyzed virulence genes related to the pathogenicity of MRSA were down-regulated at the sub-MIC concentrations of EOs, indicated that EOs could be successfully used to suppress the virulence factors and, consequently, decreased the pathogenicity of MRSA. CONCLUSION These encouraging results indicate that some of the EOs used in this study can be utilized as a natural antibiotic for the treatment of MRSA disease.
Collapse
Affiliation(s)
- Mohammad H. Gharaibeh
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030 Irbid 22110 Jordan
| | - Mohammad S. Khalifeh
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030 Irbid 22110 Jordan
| | - Esam M. Zattout
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030 Irbid 22110 Jordan
| | - Luay F. Abu-Qatouseh
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy, University of Petra, Amman, Jordan
| |
Collapse
|
238
|
Olwal CO, Ang'ienda PO, Ochiel DO. Alternative sigma factor B (σ B) and catalase enzyme contribute to Staphylococcus epidermidis biofilm's tolerance against physico-chemical disinfection. Sci Rep 2019; 9:5355. [PMID: 30926870 PMCID: PMC6440968 DOI: 10.1038/s41598-019-41797-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus epidermidis is the predominant cause of recalcitrant biofilm-associated infections, which are often highly resistant to antibiotics. Thus, the use of physico-chemical agents for disinfection offers a more effective approach to the control of S. epidermidis biofilm infections. However, the underlying tolerance mechanisms employed by S. epidermidis biofilm against these physico-chemical disinfectants remain largely unknown. The expression of a σB-dependent gene, alkaline shock protein 23 (asp23) and catalase activity by S. epidermidis biofilm and planktonic cells exposed to heat (50 °C), 0.8 M sodium chloride (NaCl), 5 mM sodium hypochlorite (NaOCl) or 50 μM hydrogen peroxide (H2O2) for 60 minutes were compared. Significantly higher asp23 expression levels were observed in biofilms exposed to 50 °C, 5 mM NaOCl or 50 μM H2O2 compared to the corresponding planktonic cells (p < 0.05). Conversely, asp23 expression levels in biofilm and planktonic cells exposed to 0.8 M NaCl were not significantly different (p > 0.05). Further, biofilms exposed to 50 °C, 0.8 M NaCl, 5 mM NaOCl or 50 μM H2O2 exhibited significantly higher catalase activity than the planktonic cells (p < 0.05). These results suggest that activities of σB and catalase may be involved in the tolerance of S. epidermidis biofilm against physico-chemical disinfection.
Collapse
|
239
|
Yang J, Qiu L, Huang Y, Chen Y, Rao S, Ruan W, Zhao G, Ye L. [The inhibition of accessory gene regulator C specific binding peptides on biofilm formation of Staphylococcus epidermidis on the surface of polyvinyl chloride in vitro]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:349-355. [PMID: 30874395 PMCID: PMC8337929 DOI: 10.7507/1002-1892.201806110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/15/2019] [Indexed: 11/03/2022]
Abstract
Objective To investigate the effect of accessory gene regulator C (agr C) specific binding peptides (named N1) on the biofilm formation of Staphylococcus epidermidis on the surface of polyvinyl chloride (PVC) materials in vitro. Methods Firstly, the two strains (ATCC35984, ATCC12228) were cultured with N1 at concentrations of 100, 200, 400, 800, and 1 600 μg/mL, respectively. The control group was cultured with agrC specific binding unrelated peptides (named N0) at the same concentrations and the absorbance ( A) value was measured after 24 hours to determine the optimal bacteriostatic concentration of N1. The two strains were cultured with N1 and N0 of the optimal concentration, respectively. The A values were measured at 6, 12, 18, 24, 30, and 48 hours to observe the effect of N1 on the biofilm formation ability of Staphylococcus epidermidis. On this basis, the surface structure of the biofilm on the surface of PVC material was observed by scanning electron microscopy after 6, 12, 18, 24, and 30 hours of incubation with PVC material sheet. The thickness of the biofilm was observed by laser confocal microscopy after 6, 12, 18, and 24 hours of incubation with ATCC35984 strain. Results The optimal bacteriostatic concentration of N1 was 800 μg/mL. ATCC 12228 strain did not form obvious biofilm after being cultured with N1 and N0. When ATCC35984 strain was cultured with N1 and N0 for 12 hours, the difference in biofilm formation ability between groups N1 and N0 was statistically significant ( P<0.05), but there was no significant difference at 6, 18, 24, 30, and 48 hours ( P>0.05). Scanning electron microscopy examination showed that mature biofilm structure was observed in ATCC35984 strain and was not observed in ATCC12228 strain. Laser confocal microscopy observation showed that the number of bacteria in the group N1 was significantly lower than that in the group N0 at 12 hours, and the most of bacteria were dead bacteria. There was no significant difference in the number of bacteria at 6, 18, and 24 hours, and the most of them were live bacteria. The biofilm thickness of group N1 was significantly lower than that of group N0 at 12 and 18 hours ( P<0.05). Conclusion The intensity of N1 inhibiting the formation of Staphylococcus epidermidis biofilm is dose-dependent. During the aggregation period, N1 can inhibit the biofilm formation by hindering the bacterial growth and aggregation. The inhibition effect on mature biofilm is not obvious.
Collapse
Affiliation(s)
- Jichen Yang
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Liangting Qiu
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Yunchao Huang
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Ya Chen
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Sunyin Rao
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Wenpeng Ruan
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Guangqiang Zhao
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118, P.R.China
| | - Lianhua Ye
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming Yunnan, 650118,
| |
Collapse
|
240
|
Effect of antibiotic infused calcium sulfate/hydroxyapatite (CAS/HA) insets on implant-associated osteitis in a femur fracture model in mice. PLoS One 2019; 14:e0213590. [PMID: 30870491 PMCID: PMC6417783 DOI: 10.1371/journal.pone.0213590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/25/2019] [Indexed: 12/19/2022] Open
Abstract
Cerament (Bonesupport Holding, Lund, Sweden) is a bioresorbable synthetic bone substitute consisting of calcium sulfate and hydroxyapatite which is successfully used as a bone graft in bone defects or in delayed and non-unions after fractures. Besides, calcium sulfate/ hydroxyapatite (CAS/HA) could have, attributed to its composition and osteoinductive properties, have great importance in the treatment of bone infections with critical size defects (CSD). Aim of the study was to evaluate the effects of antibiotic infused CAS/HA on inflammation and bone healing in an implant-associated osteitis mice model. In a standardized murine model, the left femur of 72 BALB/c mice were osteotomized, generating a CSD (2,5 mm) with stabilization through a 6-hole titanium locking plate. Osteitis has been induced through inoculation of Staphylococcus aureus (SA) into the fracture gap. To analyze the effect of CAS/HA, following groups were generated with either CAS/HA, CAS/HA with gentamycin (CAS/ HA-G) or CAS/HA with vancomycin (CAS/HA-V) insets placed into the osteotomy. Debridément and lavages were progressed on day 7 and 42 to determine the local bacterial growth and the immune reaction. Fracture healing was quantified on day 7 and 42 by x-ray and bone healing markers from blood samples. Progression of infection was assessed by estimation of colony-forming units (CFU) and immune response was analyzed by determination of Interleukin (IL)– 6 and polymorphonuclear neutrophils (PMN) in lavage samples. Osteitis induced higher IL-6 and PMN-levels in the lavage samples on day 7. Both parameters showed a reduction in all groups on day 42. CAS/HA-V revealed a significant reduction of CFU and PMNs in lavage samples on day 42. A positive effect on bone healing could only be shown in non-infected mice. Whereas, application of mere CAS/HA in infected mice did show tendencies of bone destruction and lysis, independent of impregnation with antibiotics or not. Thus, application of CAS/HA in acute implant-associated infections is not recommended. In non-infectious environments or after infect-convalescence CAS/HA could albeit serve as a suggestive tool in trauma and orthopedic surgery.
Collapse
|
241
|
Dengler Haunreiter V, Boumasmoud M, Häffner N, Wipfli D, Leimer N, Rachmühl C, Kühnert D, Achermann Y, Zbinden R, Benussi S, Vulin C, Zinkernagel AS. In-host evolution of Staphylococcus epidermidis in a pacemaker-associated endocarditis resulting in increased antibiotic tolerance. Nat Commun 2019; 10:1149. [PMID: 30850614 PMCID: PMC6408453 DOI: 10.1038/s41467-019-09053-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/13/2019] [Indexed: 02/05/2023] Open
Abstract
Treatment failure in biofilm-associated bacterial infections is an important healthcare issue. In vitro studies and mouse models suggest that bacteria enter a slow-growing/non-growing state that results in transient tolerance to antibiotics in the absence of a specific resistance mechanism. However, little clinical confirmation of antibiotic tolerant bacteria in patients exists. In this study we investigate a Staphylococcus epidermidis pacemaker-associated endocarditis, in a patient who developed a break-through bacteremia despite taking antibiotics to which the S. epidermidis isolate is fully susceptible in vitro. Characterization of the clinical S. epidermidis isolates reveals in-host evolution over the 16-week infection period, resulting in increased antibiotic tolerance of the entire population due to a prolonged lag time until growth resumption and a reduced growth rate. Furthermore, we observe adaptation towards an increased biofilm formation capacity and genetic diversification of the S. epidermidis isolates within the patient.
Collapse
Affiliation(s)
- Vanina Dengler Haunreiter
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Mathilde Boumasmoud
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Nicola Häffner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Dennis Wipfli
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Nadja Leimer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 02115, Boston, MA, USA
| | - Carole Rachmühl
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Zurich, Switzerland
| | - Denise Kühnert
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
- Max Planck Institute for the Science of Human History, 07745, Jena, Germany
| | - Yvonne Achermann
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Reinhard Zbinden
- Institute of Medical Microbiology, University of Zurich, 8006, Zurich, Switzerland
| | - Stefano Benussi
- Department of Cardiac Surgery, University Heart Center, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Clement Vulin
- Institute of Biogeochemistry and Pollutant Dynamics, ETH Zurich, 8092, Zurich, Switzerland
- Department of Environmental Microbiology, Eawag, 8600, Dübendorf, Switzerland
| | - Annelies S Zinkernagel
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland.
| |
Collapse
|
242
|
Boudjemaa R, Steenkeste K, Canette A, Briandet R, Fontaine-Aupart MP, Marlière C. Direct observation of the cell-wall remodeling in adhering Staphylococcus aureus 27217: An AFM study supported by SEM and TEM. Cell Surf 2019; 5:100018. [PMID: 32743135 PMCID: PMC7389151 DOI: 10.1016/j.tcsw.2019.100018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/19/2018] [Accepted: 01/24/2019] [Indexed: 01/28/2023] Open
Abstract
We took benefit from Atomic Force Microscopy (AFM) in the force spectroscopy mode to describe the time evolution – over 24 h – of the surface nanotopography and mechanical properties of the strain Staphylococcus aureus 27217 from bacterial adhesion to the first stage of biofilm genesis. In addition, Transmission Electron Microscopy (TEM) and Scanning Electron Microscopy (SEM) experiments allowed identifying two types of self-adhering subpopulations (the so-called “bald” and “hairy” cells) and revealed changes in their relative populations with the bacterial culture age and the protocol of preparation. We indeed observed a dramatic evanescing of the “hairy” subpopulation for samples that underwent centrifugation and resuspension processes. When examined by AFM, the “hairy” cell surface resembled to a herringbone structure characterized by upper structural units with lateral dimensions of ∼70 nm and a high Young modulus value (∼2.3 MPa), a mean depth of the trough between them of ∼15 nm and a resulting roughness of ∼5 nm. By contrast, the “bald” cells appeared much softer (∼0.35 MPa) with a roughness one order of magnitude lower. We observed too the gradual detachment of the herringbone patterns from the “hairy” bacterial envelope of cell harvested from a 16 h old culture and their progressive accumulation between the bacteria in the form of globular clusters. The secretion of a soft extracellular polymeric substance was also identified that, in addition to the globular clusters, may contribute to the initiation of the biofilm spatial organization.
Collapse
Affiliation(s)
- Rym Boudjemaa
- Institut des Sciences Moléculaires d'Orsay (ISMO), CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Karine Steenkeste
- Institut des Sciences Moléculaires d'Orsay (ISMO), CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Alexis Canette
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Institut de Biologie Paris-Seine (FR 3631), Unité Mixte de Service (UMS 30) d'Imagerie et de Cytométrie (LUMIC), Sorbonne Université, CNRS, Paris, France
| | - Romain Briandet
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Marie-Pierre Fontaine-Aupart
- Institut des Sciences Moléculaires d'Orsay (ISMO), CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Christian Marlière
- Institut des Sciences Moléculaires d'Orsay (ISMO), CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| |
Collapse
|
243
|
Transcriptional Regulation of icaADBC by both IcaR and TcaR in Staphylococcus epidermidis. J Bacteriol 2019; 201:JB.00524-18. [PMID: 30602488 DOI: 10.1128/jb.00524-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022] Open
Abstract
S. epidermidis is a primary cause of biofilm-mediated infections in humans due to adherence to foreign bodies. A major staphylococcal biofilm accumulation molecule is polysaccharide intracellular adhesin (PIA), which is synthesized by enzymes encoded by the icaADBC operon. Expression of PIA is highly variable among clinical isolates, suggesting that PIA expression levels are selected in certain niches of the host. However, the mechanisms that govern enhanced icaADBC transcription and PIA synthesis in these isolates are not known. We hypothesized that enhanced PIA synthesis in these isolates was due to function of IcaR and/or TcaR. Thus, two S. epidermidis isolates (1457 and CSF41498) with different icaADBC transcription and PIA expression levels were studied. Constitutive expression of both icaR and tcaR demonstrated that both repressors are functional and can completely repress icaADBC transcription in both 1457 and CSF41498. However, it was found that IcaR was the primary repressor for CSF41498 and TcaR was the primary repressor for 1457. Further analysis demonstrated that icaR transcription was repressed in 1457 in comparison to CSF41498, suggesting that TcaR functions as a repressor only in the absence of IcaR. Indeed, DNase I footprinting suggests IcaR and TcaR may bind to the same site within the icaR-icaA intergenic region. Lastly, we found mutants expressing variable amounts of PIA could rapidly be selected from both 1457 and CSF41498. Collectively, we propose that strains producing enhanced PIA synthesis are selected within certain niches of the host through several genetic mechanisms that function to repress icaR transcription, thus increasing PIA synthesis.IMPORTANCE Staphylococcus epidermidis is a commensal bacterium that resides on our skin. As a commensal, it protects humans from bacterial pathogens through a variety of mechanisms. However, it is also a significant cause of biofilm infections due to its ability to bind to plastic. Polysaccharide intercellular adhesin is a significant component of biofilm, and we propose that the expression of this polysaccharide is beneficial in certain host niches, such as providing extra strength when the bacterium is colonizing the lumen of a catheter, and detrimental in others, such as colonization of the skin surface. We show here that fine-tuning of icaADBC transcription, and thus PIA synthesis, is mediated via two transcriptional repressors, IcaR and TcaR.
Collapse
|
244
|
Complete Genome Sequence of Staphylococcus epidermidis CSF41498. Microbiol Resour Announc 2019; 8:MRA01138-18. [PMID: 30643875 PMCID: PMC6328648 DOI: 10.1128/mra.01138-18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/27/2018] [Indexed: 11/29/2022] Open
Abstract
Staphylococcus epidermidis CSF41498 is amenable to genetic manipulation and has been used to study mechanisms of biofilm formation. We report here the whole-genome sequence of this strain, which contains 2,427 protein-coding genes and 82 RNAs within its 2,481,008-bp-long genome, as well as three plasmids. Staphylococcus epidermidis CSF41498 is amenable to genetic manipulation and has been used to study mechanisms of biofilm formation. We report here the whole-genome sequence of this strain, which contains 2,427 protein-coding genes and 82 RNAs within its 2,481,008-bp-long genome, as well as three plasmids.
Collapse
|
245
|
LISOWSKA-ŁYSIAK KLAUDIA, KOSECKA-STROJEK MAJA, BIAŁECKA JOANNA, KASPROWICZ ANDRZEJ, GARBACZ KATARZYNA, PIECHOWICZ LIDIA, KMET VLADIMIR, SAVINI VINCENZO, MIĘDZOBRODZKI JACEK. New Insight into Genotypic and Phenotypic Relatedness of Staphylococcus aureus Strains from Human Infections or Animal Reservoirs. Pol J Microbiol 2019; 68:93-104. [PMID: 31050257 PMCID: PMC7256692 DOI: 10.21307/pjm-2019-011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/22/2018] [Accepted: 12/07/2018] [Indexed: 11/11/2022] Open
Abstract
Staphylococcus aureus is a common human and livestock opportunistic pathogen, and there is evidence of animal to human transmission. This paper aimed to recognize properties of the isolates from collections of human and livestock S. aureus strains and to estimate compatibility of results based on phenotypic tests, microarrays and the spa typing methods. The second goal was to study differences between human and animal isolates in terms of specificity of their hosts and the strain transmission among various hosts. Most strains showed multi-susceptible profiles and produced enzymes on a high level, and they were phenotypically and genetically similar. However, in contrast to the Polish bovine mastitis strains, the Slovakian strains were multi-resistant. In this research, the strains showed significant differences in terms of their phenotypic manifestations and the presence of hemolysins genes; however, other enzyme-encoding genes correlated to a higher extent with the microarrays results. Interestingly, there was a lack of enterotoxin genes in human Poultry-like protein A+ strains in comparison to other human strains. Our study showed that differences between virulence profiles of the human and animal strains correlated with their origin rather than their hosts, and any trait allowed clearly distinguishing between them based on the microarray results. Staphylococcus aureus is a common human and livestock opportunistic pathogen, and there is evidence of animal to human transmission. This paper aimed to recognize properties of the isolates from collections of human and livestock S. aureus strains and to estimate compatibility of results based on phenotypic tests, microarrays and the spa typing methods. The second goal was to study differences between human and animal isolates in terms of specificity of their hosts and the strain transmission among various hosts. Most strains showed multi-susceptible profiles and produced enzymes on a high level, and they were phenotypically and genetically similar. However, in contrast to the Polish bovine mastitis strains, the Slovakian strains were multi-resistant. In this research, the strains showed significant differences in terms of their phenotypic manifestations and the presence of hemolysins genes; however, other enzyme-encoding genes correlated to a higher extent with the microarrays results. Interestingly, there was a lack of enterotoxin genes in human Poultry-like protein A+ strains in comparison to other human strains. Our study showed that differences between virulence profiles of the human and animal strains correlated with their origin rather than their hosts, and any trait allowed clearly distinguishing between them based on the microarray results.
Collapse
Affiliation(s)
- KLAUDIA LISOWSKA-ŁYSIAK
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - MAJA KOSECKA-STROJEK
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - JOANNA BIAŁECKA
- Centre of Microbiological Research and Autovaccines, Krakow, Poland
| | | | - KATARZYNA GARBACZ
- Department of Oral Microbiology, Medical Faculty, Medical University of Gdansk, Gdansk, Poland
| | - LIDIA PIECHOWICZ
- Department of Medical Microbiology, Medical Faculty, Medical University of Gdansk, Gdansk, Poland
| | - VLADIMIR KMET
- Centre of Biosciences, Institute of Animal Physiology, Slovak Academy of Sciences, Slovakia
| | - VINCENZO SAVINI
- Clinical Microbiology and Virology, Spirito Santo Hospital, Pescara (PE), Italy
| | - JACEK MIĘDZOBRODZKI
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
246
|
|
247
|
Bai JR, Wu YP, Elena G, Zhong K, Gao H. Insight into the effect of quinic acid on biofilm formed byStaphylococcus aureus. RSC Adv 2019; 9:3938-3945. [PMID: 35518066 PMCID: PMC9060517 DOI: 10.1039/c8ra09136f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/22/2019] [Indexed: 01/08/2023] Open
Abstract
The biofilm formation of Staphylococcus aureus on food contact surfaces is the main risk of food contamination. In the present study, we firstly investigated the inhibitory effect of quinic acid (QA) on biofilm formed by S. aureus. Crystal violet staining assay and microscopy analysis clearly showed that QA at sub-MIC concentrations was able to significantly reduce the biofilm biomass and cause a collapse on biofilm architecture. Meanwhile, fibrinogen binding assay showed that QA had obviously effect on the S. aureus bacteria adhesion. XTT reduction assay and confocal laser scanning microscopic images revealed that QA significantly decreased metabolic activity and viability of biofilm cells. In addition, qRT-PCR analysis explored the potential inhibitory mechanism of QA against biofilm formation, which indicated that QA significantly repressed the gene sarA and activated the gene agrA. Moreover, QA exhibited a highly ability to reduce the number of sessile S. aureus cells adhered on the stainless steel. So, it was suggested that QA could be used as a promising antibiofilm agent to control biofilm formation of S. aureus. QA effectively inhibited S. aureus biofilm formation. The key genes of biofilm inhibition induced by QA were agrA and sarA.![]()
Collapse
Affiliation(s)
- Jin-Rong Bai
- College of Light Industry
- Textile and Food Engineering and Healthy Food Evaluation Research Center
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Yan-Ping Wu
- College of Light Industry
- Textile and Food Engineering and Healthy Food Evaluation Research Center
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Grosu Elena
- College of Light Industry
- Textile and Food Engineering and Healthy Food Evaluation Research Center
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Kai Zhong
- College of Light Industry
- Textile and Food Engineering and Healthy Food Evaluation Research Center
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Hong Gao
- College of Light Industry
- Textile and Food Engineering and Healthy Food Evaluation Research Center
- Sichuan University
- Chengdu 610065
- P. R. China
| |
Collapse
|
248
|
Naik BI, Roger C, Ikeda K, Todorovic MS, Wallis SC, Lipman J, Roberts JA. Comparative total and unbound pharmacokinetics of cefazolin administered by bolus versus continuous infusion in patients undergoing major surgery: a randomized controlled trial. Br J Anaesth 2018; 118:876-882. [PMID: 28505360 DOI: 10.1093/bja/aex026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2017] [Indexed: 01/04/2023] Open
Abstract
Background. Perioperative administration of cefazolin reduces the incidence of perioperative infections. Intraoperative re-dosing of cefazolin is commonly given between 2 and 5 h after the initial dose. This study was undertaken to determine whether intraoperative continuous infusions of cefazolin achieve better probability of target attainment (PTA) and fractional target attainment (FTA) than intermittent dosing. Methods. Patients undergoing major surgery received cefazolin 2 g before surgical incision. They were subsequently randomized to receive either an intermittent bolus (2 g every 4 h) or continuous infusion (500 mg h -1 ) of cefazolin until skin closure. Blood samples were analysed for total and unbound cefazolin concentrations using a validated chromatographic method. Population pharmacokinetic modelling was performed using Pmetrics ® software. Calculations of PTA and FTA were performed for common pathogens. Results. Ten patients were enrolled in each arm. A two-compartment linear model best described the time course of the total plasma cefazolin concentrations. The covariates that improved the model were body weight and creatinine clearance. Protein binding varied with time [mean (range) 69 (44-80)%] with a fixed 21% unbound value of cefazolin used for the simulations (120 min post-initial dosing). Mean ( sd ) central volume of distribution was 5.73 (2.42) litres, and total cefazolin clearance was 4.72 (1.1) litres h -1 . Continuous infusions of cefazolin consistently achieved better drug exposures and FTA for different weight and creatinine clearances, particularly for less susceptible pathogens. Conclusions. Our study demonstrates that intraoperative continuous infusions of cefazolin increase the achievement of target plasma concentrations, even with lower infusion doses. Renal function and body weight are important when considering the need for alternative dosing regimens. Clinical trial registration. NCT02058979.
Collapse
Affiliation(s)
- B I Naik
- Department of Anesthesiology, PO Box 800710, Charlottesville, VA, 22908, USA.,Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - C Roger
- Burns, Trauma and Critical Care Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - K Ikeda
- Department of Anesthesiology, PO Box 800710, Charlottesville, VA, 22908, USA
| | - M S Todorovic
- Department of Anesthesiology, Washington University, St Louis, MO, USA
| | - S C Wallis
- Burns, Trauma and Critical Care Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - J Lipman
- Burns, Trauma and Critical Care Research Centre, The University of Queensland, Brisbane, Queensland, Australia.,Department of Intensive Care Medicine
| | - J A Roberts
- Burns, Trauma and Critical Care Research Centre, The University of Queensland, Brisbane, Queensland, Australia.,Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital.,Centre for Translational Anti-infective Pharmacodynamics, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
249
|
Khatoon Z, McTiernan CD, Suuronen EJ, Mah TF, Alarcon EI. Bacterial biofilm formation on implantable devices and approaches to its treatment and prevention. Heliyon 2018; 4:e01067. [PMID: 30619958 PMCID: PMC6312881 DOI: 10.1016/j.heliyon.2018.e01067] [Citation(s) in RCA: 595] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
In living organisms, biofilms are defined as complex communities of bacteria residing within an exopolysaccharide matrix that adheres to a surface. In the clinic, they are typically the cause of chronic, nosocomial, and medical device-related infections. Due to the antibiotic-resistant nature of biofilms, the use of antibiotics alone is ineffective for treating biofilm-related infections. In this review, we present a brief overview of concepts of bacterial biofilm formation, and current state-of-the-art therapeutic approaches for preventing and treating biofilms. Also, we have reviewed the prevalence of such infections on medical devices and discussed the future challenges that need to be overcome in order to successfully treat biofilms using the novel technologies being developed.
Collapse
Affiliation(s)
- Zohra Khatoon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
| | - Christopher D. McTiernan
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
| | - Erik J. Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
| | - Thien-Fah Mah
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, K1Y 4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
250
|
Manara S, Pasolli E, Dolce D, Ravenni N, Campana S, Armanini F, Asnicar F, Mengoni A, Galli L, Montagnani C, Venturini E, Rota-Stabelli O, Grandi G, Taccetti G, Segata N. Whole-genome epidemiology, characterisation, and phylogenetic reconstruction of Staphylococcus aureus strains in a paediatric hospital. Genome Med 2018; 10:82. [PMID: 30424799 PMCID: PMC6234625 DOI: 10.1186/s13073-018-0593-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is an opportunistic pathogen and a leading cause of nosocomial infections. It can acquire resistance to all the antibiotics that entered the clinics to date, and the World Health Organization defined it as a high-priority pathogen for research and development of new antibiotics. A deeper understanding of the genetic variability of S. aureus in clinical settings would lead to a better comprehension of its pathogenic potential and improved strategies to contrast its virulence and resistance. However, the number of comprehensive studies addressing clinical cohorts of S. aureus infections by simultaneously looking at the epidemiology, phylogenetic reconstruction, genomic characterisation, and transmission pathways of infective clones is currently low, thus limiting global surveillance and epidemiological monitoring. METHODS We applied whole-genome shotgun sequencing (WGS) to 184 S. aureus isolates from 135 patients treated in different operative units of an Italian paediatric hospital over a timespan of 3 years, including both methicillin-resistant S. aureus (MRSA) and methicillin-sensitive S. aureus (MSSA) from different infection types. We typed known and unknown clones from their genomes by multilocus sequence typing (MLST), Staphylococcal Cassette Chromosome mec (SCCmec), Staphylococcal protein A gene (spa), and Panton-Valentine Leukocidin (PVL), and we inferred their whole-genome phylogeny. We explored the prevalence of virulence and antibiotic resistance genes in our cohort, and the conservation of genes encoding vaccine candidates. We also performed a timed phylogenetic investigation for a potential outbreak of a newly emerging nosocomial clone. RESULTS The phylogeny of the 135 single-patient S. aureus isolates showed a high level of diversity, including 80 different lineages, and co-presence of local, global, livestock-associated, and hypervirulent clones. Five of these clones do not have representative genomes in public databases. Variability in the epidemiology is mirrored by variability in the SCCmec cassettes, with some novel variants of the type IV cassette carrying extra antibiotic resistances. Virulence and resistance genes were unevenly distributed across different clones and infection types, with highly resistant and lowly virulent clones showing strong association with chronic diseases, and highly virulent strains only reported in acute infections. Antigens included in vaccine formulations undergoing clinical trials were conserved at different levels in our cohort, with only a few highly prevalent genes fully conserved, potentially explaining the difficulty of developing a vaccine against S. aureus. We also found a recently diverged ST1-SCCmecIV-t127 PVL- clone suspected to be hospital-specific, but time-resolved integrative phylogenetic analysis refuted this hypothesis and suggested that this quickly emerging lineage was acquired independently by patients. CONCLUSIONS Whole genome sequencing allowed us to study the epidemiology and genomic repertoire of S. aureus in a clinical setting and provided evidence of its often underestimated complexity. Some virulence factors and clones are specific of disease types, but the variability and dispensability of many antigens considered for vaccine development together with the quickly changing epidemiology of S. aureus makes it very challenging to develop full-coverage therapies and vaccines. Expanding WGS-based surveillance of S. aureus to many more hospitals would allow the identification of specific strains representing the main burden of infection and therefore reassessing the efforts for the discovery of new treatments and clinical practices.
Collapse
Affiliation(s)
- Serena Manara
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Edoardo Pasolli
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Daniela Dolce
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Novella Ravenni
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Silvia Campana
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | | | | | - Alessio Mengoni
- Department of Biology, University of Florence, Florence, Italy
| | - Luisa Galli
- Department of Health Sciences, University of Florence, Florence, Italy
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Carlotta Montagnani
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Elisabetta Venturini
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Omar Rota-Stabelli
- Department of Sustainable Agro-Ecosystems and Bioresources, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Guido Grandi
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Giovanni Taccetti
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Nicola Segata
- Centre for Integrative Biology, University of Trento, Trento, Italy.
| |
Collapse
|