51
|
Leanse LG, Zeng X, Dai T. Potentiated antimicrobial blue light killing of methicillin resistant Staphylococcus aureus by pyocyanin. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 215:112109. [PMID: 33486397 DOI: 10.1016/j.jphotobiol.2020.112109] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/11/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022]
Abstract
As antimicrobial resistance continues to threaten the efficacy of conventional antibiotic therapy, it is paramount that we investigate innovative approaches to treat infectious diseases. In this study, we investigated the antimicrobial capabilities of the innovative combination of antimicrobial blue light (aBL; 405 nm wavelength) with the Pseudomonas aeruginosa pigment pyocyanin against methicillin resistant Staphylococcus aureus (MSRA. We explored the effects of different radiant exposures of aBL and increasing concentrations of pyocyanin against planktonic cells and those within biofilms. In addition, we investigated the effect of the aBL/pyocyanin on the endogenous staphyloxanthin pigment, as well as the role of hydrogen peroxide and singlet oxygen scavenging in the efficacy of this combination. Lastly, we investigated the potential for the aBL/pyocyanin to reduce the MRSA burden within a proof-of-principle mouse abrasion infection model. We found pyocyanin to be a powerful potentiator of aBL activity under all in vitro conditions tested. In addition, we serendipitously discovered the capability of the aBL/pyocyanin combination to bleach staphyloxanthin within colonies of MRSA. Furthermore, we established that singlet oxygen is an important mediator during combined aBL/pyocyanin exposure. Moreover, we found that the combination of aBL and pyocyanin could significantly reduce the viability of MRSA within a proof-of-principle early onset MRSA skin abrasion infection. Exposure to the treatment did not have deleterious effects on skin tissue. In conclusion, the combination of aBL and pyocyanin represents a potentially powerful therapeutic modality for the treatment of infections caused by MRSA.
Collapse
Affiliation(s)
- Leon G Leanse
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Xiaojing Zeng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; School of Medicine, Shanghai Jiao Tong University, China
| | - Tianhong Dai
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| |
Collapse
|
52
|
Li P, Gao Z, Tan Z, Xiao J, Wei L, Chen Y. New developments in anti-biofilm intervention towards effective management of orthopedic device related infections (ODRI's). BIOFOULING 2021; 37:1-35. [PMID: 33618584 DOI: 10.1080/08927014.2020.1869725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 06/12/2023]
Abstract
Orthopedic device related infections (ODRI's) represent a difficult to treat situation owing to their biofilm based nature. Biofilm infections once established are difficult to eradicate even with an aggressive treatment regimen due to their recalcitrance towards antibiotics and immune attack. The involvement of antibiotic resistant pathogens as the etiological agent further worsens the overall clinical picture, pressing on the need to look into alternative treatment strategies. The present review highlightes the microbiological challenges associated with treatment of ODRI's due to biofilm formation on the implant surface. Further, it details the newer anti-infective modalities that work either by preventing biofilm formation and/or through effective disruption of the mature biofilms formed on the medical implant. The study, therefore aims to provide a comprehensive insight into the newer anti-biofilm interventions (non-antibiotic approaches) and a better understanding of their mechanism of action essential for improved management of orthopedic implant infections.
Collapse
Affiliation(s)
- Ping Li
- Department of Orthopedics, Ya'an People's Hospital, Yaan City, China
| | - Zhenwu Gao
- Department of Orthopedics, Shanxi Bethune Hospital, Taiyuan City, China
| | - Zhenwei Tan
- Department of Orthopedics, Western Theater Air Force Hospital of PLA, Chengdu, China
| | - Jun Xiao
- Department of Orthopedics, Ya'an People's Hospital, Yaan City, China
| | - Li Wei
- Nursing Department, Three Gorges Hospital Affiliated to Chongqing University, Chongqing, China
| | - Yirui Chen
- Department of Orthopedics, Three Gorges Hospital Affiliated to Chongqing University, Chongqing, China
| |
Collapse
|
53
|
Cai YM. Non-surface Attached Bacterial Aggregates: A Ubiquitous Third Lifestyle. Front Microbiol 2020; 11:557035. [PMID: 33343514 PMCID: PMC7746683 DOI: 10.3389/fmicb.2020.557035] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/13/2020] [Indexed: 01/03/2023] Open
Abstract
Bacteria are now generally believed to adopt two main lifestyles: planktonic individuals, or surface-attached biofilms. However, in recent years medical microbiologists started to stress that suspended bacterial aggregates are a major form of bacterial communities in chronic infection sites. Despite sharing many similarities with surface-attached biofilms and are thus generally defined as biofilm-like aggregates, these non-attached clumps of cells in vivo show much smaller sizes and different formation mechanisms. Furthermore, ex vivo clinical isolates were frequently reported to be less attached to abiotic surfaces when compared to standard type strains. While this third lifestyle is starting to draw heavy attention in clinical studies, it has a long history in natural and environmental sciences. For example, marine gel particles formed by bacteria attachment to phytoplankton exopolymers have been well documented in oceans; large river and lake snows loaded with bacterial aggregates are frequently found in freshwater systems; multispecies bacterial "flocs" have long been used in wastewater treatment. This review focuses on non-attached aggregates found in a variety of natural and clinical settings, as well as some recent technical developments facilitating aggregate research. The aim is to summarise the characteristics of different types of bacterial aggregates, bridging the knowledge gap, provoking new perspectives for researchers from different fields, and highlighting the importance of more research input in this third lifestyle of bacteria closely relevant to our daily life.
Collapse
Affiliation(s)
- Yu-Ming Cai
- National Biofilms Innovation Centre, Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- Biological Sciences, Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
54
|
Three-Dimensional In Vitro Staphylococcus aureus Abscess Communities Display Antibiotic Tolerance and Protection from Neutrophil Clearance. Infect Immun 2020; 88:IAI.00293-20. [PMID: 32817328 DOI: 10.1128/iai.00293-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a prominent human pathogen in bone and soft-tissue infections. Pathophysiology involves abscess formation, which consists of central staphylococcal abscess communities (SACs), surrounded by a fibrin pseudocapsule and infiltrating immune cells. Protection against the ingress of immune cells such as neutrophils, or tolerance to antibiotics, remains largely unknown for SACs and is limited by the lack of availability of in vitro models. We describe a three-dimensional in vitro model of SACs grown in a human plasma-supplemented collagen gel. The in vitro SACs reached their maximum size by 24 h and elaborated a fibrin pseudocapsule, as confirmed by electron and immunofluorescence microscopy. The in vitro SACs tolerated 100× the MIC of gentamicin alone and in combination with rifampin, while planktonic controls and mechanically dispersed SACs were efficiently killed. To simulate a host response, SACs were exposed to differentiated PLB-985 neutrophil-like (dPLB) cells and to primary human neutrophils at an early stage of SAC formation or after maturation at 24 h. Both cell types were unable to clear mature in vitro SACs, but dPLB cells prevented SAC growth upon early exposure before pseudocapsule maturation. Neutrophil exposure after plasmin pretreatment of the SACs resulted in a significant decrease in the number of bacteria within the SACs. The in vitro SAC model mimics key in vivo features, offers a new tool to study host-pathogen interactions and drug efficacy assessment, and has revealed the functionality of the S. aureus pseudocapsule in protecting the bacteria from host phagocytic responses and antibiotics.
Collapse
|
55
|
Staphylococcus aureus Aggregates on Orthopedic Materials under Varying Levels of Shear Stress. Appl Environ Microbiol 2020; 86:AEM.01234-20. [PMID: 32709721 DOI: 10.1128/aem.01234-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/16/2020] [Indexed: 02/08/2023] Open
Abstract
Periprosthetic joint infection (PJI) occurring after artificial joint replacement is a major clinical issue requiring multiple surgeries and antibiotic interventions. Staphylococcus aureus is the bacterium most commonly responsible for PJI. Recent in vitro research has shown that staphylococcal strains rapidly form aggregates in the presence of synovial fluid (SF). We hypothesize that these aggregates provide early protection to bacteria entering the wound site, allowing them time to attach to the implant surface, leading to biofilm formation. Thus, understanding the attachment kinetics of these aggregates is critical in understanding their adhesion to various biomaterial surfaces. In this study, the number, size, and surface area coverage of aggregates as well as of single cells of S. aureus were quantified under various conditions on different orthopedic materials relevant to orthopedic surgery: stainless steel (316L), titanium (Ti), hydroxyapatite (HA), and polyethylene (PE). It was observed that, regardless of the material type, SF-induced aggregation resulted in reduced aggregate surface attachment and greater aggregate size than the single-cell populations under various shear stresses. Additionally, the surface area coverage of bacterial aggregates on PE was relatively high compared to that on other materials, which could potentially be due to the rougher surface of PE. Furthermore, increasing shear stress to 78 mPa decreased aggregate attachment to Ti and HA while increasing the aggregates' average size. Therefore, this study demonstrates that SF induced inhibition of aggregate attachment to all materials, suggesting that biofilm formation is initiated by lodging of aggregates on the surface features of implants and host tissues.IMPORTANCE Periprosthetic joint infection occurring after artificial joint replacement is a major clinical issue that require repeated surgeries and antibiotic interventions. Unfortunately, 26% of patients die within 5 years of developing these infections. Staphylococcus aureus is the bacterium most commonly responsible for this problem and can form biofilms to provide protection from antibiotics as well as the immune system. Although biofilms are evident on the infected implants, it is unclear how these are attached to the surface in the first place. Recent in vitro investigations have shown that staphylococcal strains rapidly form aggregates in the presence of synovial fluid and provide protection to bacteria, thus allowing them time to attach to the implant surface, leading to biofilm formation. In this study, we investigated the attachment kinetics of Staphylococcus aureus aggregates on different orthopedic materials. The information presented in this article will be useful in surgical management and implant design.
Collapse
|
56
|
Franceschini M, Sandiford NA, Cerbone V, Araujo LCTD, Kendoff D. Defensive antibacterial coating in revision total hip arthroplasty: new concept and early experience. Hip Int 2020; 30:7-11. [PMID: 32907424 DOI: 10.1177/1120700020917125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Infections remains the most feared complication in total hip arthroplasty (THA). New strategies of PJI prevention includes coating of conventional implants. Defensive Antibacterial Coating (DAC), an antibacterial hydrogel coating made of hyaluronan, poly-D and L-lactide can protect biomaterials as an effective barrier at the time of implantation. In addition, it can be used with topical antibiotics to prevent early colonisation of the implant. SCOPE This manuscript describes the detailed function of the DAC in general as well as an analysis of its use in revision THA in a series of 28 patients in a short-term follow-up.Its use in patients undergoing cementless re-implantation after 2-staged procedures in THA is described in detail within the manuscript. CONCLUSION DAC found to be effective in terms of infection control and safety in our patient cohort and has been expanded for cementless 1-staged revisions in PJI of the hip in our institution.
Collapse
|
57
|
Liu T, Luo J, Bi G, Du Z, Kong J, Chen Y. Antibacterial synergy between linezolid and baicalein against methicillin-resistant Staphylococcus aureus biofilm in vivo. Microb Pathog 2020; 147:104411. [PMID: 32745664 DOI: 10.1016/j.micpath.2020.104411] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/09/2020] [Accepted: 07/18/2020] [Indexed: 10/23/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) can form biofilms, which prevents the penetration of antibiotics, decreasing their efficacy. This study investigated whether baicalein has synergistic antibacterial effects with linezolid in vivo. We cultivated MRSA 17546 biofilms on silicone implants and inserted them into the air pouches of rat models. The rats were treated with linezolid, baicalein, or a combination therapy for three consecutive days. All treatments reduced the number of colony-forming units (CFU) in the biofilms compared to the control (p < 0.05). However, by day two, the CFU counts were significantly lower in the combination group than in the individual treatment groups (p < 0.05). Histological analysis of the air pouches showed that the severity of the inflammatory cell infiltration was severe in the combination therapy group. In the combination group, the biofilm structure on the implant's surface was sparse and more free colonies could be seen by scanning electron microscopy (SEM); by day three, no obvious biofilm was observed. The serum levels of Staphylococcus enterotoxin A (SEA), C-reactive protein (CRP), and procalcitonin (PCT) were the lowest in the group where rats were treated with the combination of baicalein and linezolid (p < 0.05) compared to other groups. The results suggest that baicalein may inhibit the accessory gene regulator system, reducing the expression of SEA, thus lowering CRP and PCT levels. Furthermore, the inhibitory effect was more pronounced when baicalein was combined with linezolid. These results provide an important basis for the development of a new combination regimen to treat patients with biofilm-associated MRSA infections.
Collapse
Affiliation(s)
- Tangjuan Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Jing Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Guan Bi
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Zhongye Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Jinliang Kong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.
| | - Yiqiang Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.
| |
Collapse
|
58
|
Bidossi A, Bottagisio M, Savadori P, De Vecchi E. Identification and Characterization of Planktonic Biofilm-Like Aggregates in Infected Synovial Fluids From Joint Infections. Front Microbiol 2020; 11:1368. [PMID: 32714301 PMCID: PMC7344156 DOI: 10.3389/fmicb.2020.01368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/27/2020] [Indexed: 01/10/2023] Open
Abstract
Recent in vitro studies reported the exceptional ability of some bacterial species to form biofilm-like aggregates in human and animal synovial fluids (SF), but evidences from infected clinical samples are still lacking. In this study, we investigated whether this bacterial phenotype was present in infected SFs collected from joint infections and if it was maintained in in vitro settings. SFs sent for culture to the Laboratory of Microbiology of our institute were directly analyzed by means of confocal laser scanning microscopy (CLSM), and the infective agents were isolated for further in vitro tests. Moreover, sterile SF was collected from patients who did not receive previous antibiotic therapy to investigate the formation of bacterial aggregates, together with biofilm and matrix production on a titanium surface. Finally, antibiotic susceptibility studies were performed by using bovine SF. Four Staphylococcus aureus, one Staphylococcus lugdunensis, and one Prevotella bivia strain were identified in the infected SFs. The CLSM analysis showed that all staphylococci were present as a mixture of single cells and bacterial clumps surrounded by an exopolymeric substance, which comprised SF-derived fibrin, while all P. bivia cells appeared separated. Despite that, differences in the ability to aggregate between S. aureus and S. lugdunensis were observed in clinical SFs. These different phenotypes were further confirmed by in vitro growth, even though the application of such ex vivo approach lead all staphylococci to form exceptionally large microbial aggregates, which are several folds bigger than those observed in clinical samples. Planktonic aggregates challenged for antibiotic susceptibility revealed a sharp increase of recalcitrance to the treatments. Although this is still at a preliminary stage, the present work confirmed the ability of staphylococci to form free-floating biofilm-like aggregates in infected SF from patients with joint infections. Furthermore, the obtained results pointed out that future in vitro research on joint infections will benefit from the use of human- or animal-derived SF. Even though this approach should be carefully validated in further studies comprising a larger microbial population, these findings pose new challenges in the treatment of infected native and prosthetic joints and for the approach to new investigations.
Collapse
Affiliation(s)
- Alessandro Bidossi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Marta Bottagisio
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Paolo Savadori
- Department of Endodontics, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Elena De Vecchi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
59
|
Leanse LG, Goh XS, Cheng JX, Hooper DC, Dai T. Dual-wavelength photo-killing of methicillin-resistant Staphylococcus aureus. JCI Insight 2020; 5:134343. [PMID: 32493838 DOI: 10.1172/jci.insight.134343] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/23/2020] [Indexed: 12/20/2022] Open
Abstract
With the effectiveness of antimicrobials declining as antimicrobial resistance continues to threaten public health, we must look to alternative strategies for the treatment of infections. In this study, we investigated an innovative, drug-free, dual-wavelength irradiation approach that combines 2 wavelengths of light, 460 nm and 405 nm, against methicillin-resistant Staphylococcus aureus (MRSA). MRSA was initially irradiated with 460-nm light (90-360 J/cm2) and subsequently irradiated with aliquots of 405-nm light (54-324 J/cm2). For in vivo studies, mouse skin was abraded and infected with approximately 107 CFUs of MRSA and incubated for 3 hours before irradiating with 460 nm (360 J/cm2) and 405 nm (342 J/cm2). Naive mouse skin was also irradiated to investigate apoptosis. We found that staphyloxanthin, the carotenoid pigment in MRSA cells, promoted resistance to the antimicrobial effects of 405-nm light. In addition, we found that the photolytic effect of 460-nm light on staphyloxanthin attenuated resistance of MRSA to 405-nm light killing. Irradiation of 460 nm alone did not elicit any antimicrobial effect on MRSA. In a proof-of-principle mouse skin abrasion infection model, we observed significant killing of MRSA using the dual-wavelength irradiation approach. However, when either wavelength of light was administered alone, no significant decrease in bacterial viability was observed. Moreover, exposure of the dual-wavelength irradiation to naive mouse skin did not result in any visible apoptosis. In conclusion, a dual-wavelength irradiation strategy may offer an innovative, effective, and safe approach for the treatment of skin infections caused by MRSA.
Collapse
Affiliation(s)
- Leon G Leanse
- Wellman Center for Photomedicine.,Vaccine and Immunotherapy Center, and
| | | | - Ji-Xin Cheng
- Department of Chemistry, Boston University, Boston, Massachusetts, USA
| | - David C Hooper
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tianhong Dai
- Wellman Center for Photomedicine.,Vaccine and Immunotherapy Center, and
| |
Collapse
|
60
|
Gilbertie JM, Schaer TP, Schubert AG, Jacob ME, Menegatti S, Lavoie RA, Schnabel LV. Platelet-rich plasma lysate displays antibiofilm properties and restores antimicrobial activity against synovial fluid biofilms in vitro. J Orthop Res 2020; 38:1365-1374. [PMID: 31922274 PMCID: PMC8018705 DOI: 10.1002/jor.24584] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/06/2019] [Accepted: 01/03/2020] [Indexed: 02/04/2023]
Abstract
Infectious arthritis is difficult to treat in both human and veterinary clinical practice. Recent literature reports Staphylococcus aureus as well as other gram-positive and gram-negative isolates forming free-floating biofilms in both human and equine synovial fluid that are tolerant to traditional antimicrobial therapy. Using an in vitro equine model, we investigated the ability of platelet-rich plasma (PRP) formulations to combat synovial fluid biofilm aggregates. Synovial fluid was infected, and biofilm aggregates allowed to form over a 2-hour period. PRP was collected and processed into different formulations by platelet concentration, leukocyte presence, and activation or lysis. Infected synovial fluid was treated with different PRP formulations with or without aminoglycoside cotreatment. Bacterial load (colony-forming unit/mL) was determined by serial dilutions and plate counting at 8 hours posttreatment. All PRP formulations displayed antimicrobial properties; however, formulations containing higher concentrations of platelets without leukocytes had increased antimicrobial activity. Lysis of PRP and pooling of the PRP lysate (PRP-L) from multiple horses as compared to individual horses further increased antimicrobial activity. This activity was lost with the removal of the plasma component or inhibition of the proteolytic activity within the plasma. Fractionation of pooled PRP-L identified the bioactive components to be cationic and low-molecular weight (<10 kDa). Overall, PRP-L exhibited synergism with amikacin against aminoglycoside tolerant biofilm aggregates with greater activity against gram-positive bacteria. In conclusion, the use of PRP-L has the potential to augment current antimicrobial treatment regimens which could lead to a decrease in morbidity and mortality associated with infectious arthritis.
Collapse
Affiliation(s)
- Jessica M. Gilbertie
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina,Department of Clinical Studies New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Thomas P. Schaer
- Department of Clinical Studies New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Alicia G. Schubert
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Megan E. Jacob
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina,Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Stefano Menegatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - R. Ashton Lavoie
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
61
|
Kuehl R, Morata L, Meylan S, Mensa J, Soriano A. When antibiotics fail: a clinical and microbiological perspective on antibiotic tolerance and persistence of Staphylococcus aureus. J Antimicrob Chemother 2020; 75:1071-1086. [PMID: 32016348 DOI: 10.1093/jac/dkz559] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen causing a vast array of infections with significant mortality. Its versatile physiology enables it to adapt to various environments. Specific physiological changes are thought to underlie the frequent failure of antimicrobial therapy despite susceptibility in standard microbiological assays. Bacteria capable of surviving high antibiotic concentrations despite having a genetically susceptible background are described as 'antibiotic tolerant'. In this review, we put current knowledge on environmental triggers and molecular mechanisms of increased antibiotic survival of S. aureus into its clinical context. We discuss animal and clinical evidence of its significance and outline strategies to overcome infections with antibiotic-tolerant S. aureus.
Collapse
Affiliation(s)
- Richard Kuehl
- Service of Infectious Diseases, Hospital Clinic of Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Laura Morata
- Service of Infectious Diseases, Hospital Clinic of Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Sylvain Meylan
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
- Division de Maladies Infectieuses, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Josep Mensa
- Service of Infectious Diseases, Hospital Clinic of Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Alex Soriano
- Service of Infectious Diseases, Hospital Clinic of Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| |
Collapse
|
62
|
Pestrak MJ, Gupta TT, Dusane DH, Guzior DV, Staats A, Harro J, Horswill AR, Stoodley P. Investigation of synovial fluid induced Staphylococcus aureus aggregate development and its impact on surface attachment and biofilm formation. PLoS One 2020; 15:e0231791. [PMID: 32302361 PMCID: PMC7164621 DOI: 10.1371/journal.pone.0231791] [Citation(s) in RCA: 32] [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: 01/21/2020] [Accepted: 03/31/2020] [Indexed: 12/01/2022] Open
Abstract
Periprosthetic joint infections (PJIs) are a devastating complication that occurs in 2% of patients following joint replacement. These infections are costly and difficult to treat, often requiring multiple corrective surgeries and prolonged antimicrobial treatments. The Gram-positive bacterium Staphylococcus aureus is one of the most common causes of PJIs, and it is often resistant to a number of commonly used antimicrobials. This tolerance can be partially attributed to the ability of S. aureus to form biofilms. Biofilms associated with the surface of indwelling medical devices have been observed on components removed during chronic infection, however, the development and localization of biofilms during PJIs remains unclear. Prior studies have demonstrated that synovial fluid, in the joint cavity, promotes the development of bacterial aggregates with many biofilm-like properties, including antibiotic resistance. We anticipate these aggregates have an important role in biofilm formation and antibiotic tolerance during PJIs. Therefore, we sought to determine specifically how synovial fluid promotes aggregate formation and the impact of this process on surface attachment. Using flow cytometry and microscopy, we quantified the aggregation of various clinical S. aureus strains following exposure to purified synovial fluid components. We determined that fibrinogen and fibronectin promoted bacterial aggregation, while cell free DNA, serum albumin, and hyaluronic acid had minimal effect. To determine how synovial fluid mediated aggregation affects surface attachment, we utilized microscopy to measure bacterial attachment. Surprisingly, we found that synovial fluid significantly impeded bacterial surface attachment to a variety of materials. We conclude from this study that fibrinogen and fibronectin in synovial fluid have a crucial role in promoting bacterial aggregation and inhibiting surface adhesion during PJI. Collectively, we propose that synovial fluid may have conflicting protective roles for the host by preventing adhesion to surfaces, but by promoting bacterial aggregation is also contributing to the development of antibiotic tolerance.
Collapse
Affiliation(s)
- Matthew J. Pestrak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Tripti Thapa Gupta
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Devendra H. Dusane
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Doug V. Guzior
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Amelia Staats
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Jan Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, United States of America
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
- Department of Orthopedics, The Ohio State University, Columbus, Ohio, United States of America
- National Centre for Advanced Tribology at Southampton (nCATS) and National Biofilm Innovation Centre (NBIC), Mechanical Engineering, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
63
|
Delaney LJ, Basgul C, MacDonald DW, Fitzgerald K, Hickok NJ, Kurtz SM, Forsberg F. Acoustic Parameters for Optimal Ultrasound-Triggered Release from Novel Spinal Hardware Devices. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:350-358. [PMID: 31732196 PMCID: PMC7139856 DOI: 10.1016/j.ultrasmedbio.2019.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/28/2019] [Accepted: 10/01/2019] [Indexed: 05/06/2023]
Abstract
Post-operative infection is a catastrophic complication of spinal fusion surgery, with rates as high as 10%, and existing preventative measures (i.e., peri-operative antibiotics) are only partially successful. To combat this clinical problem, we have designed a drug delivery system around polyether ether ketone clips to be used for prophylactic post-surgical release of antibiotics upon application of ultrasound. The overall hypothesis is that antimicrobial release from this system will aggressively combat post-surgical bacterial survival. This study investigated a set of acoustic parameters optimized for in vitro ultrasound-triggered coating rupture and subsequent release of encapsulated prophylactic antibiotics. We determined that a transducer frequency of 1.7 MHz produced the most consistent burst release and that, at this frequency, a pulse repetition frequency of 6.4 kHz and acoustic output power of 100% (3.41 MPa) produced the greatest release, representing an important proof of principle and the basis for continued development of this novel drug delivery system.
Collapse
Affiliation(s)
- Lauren J Delaney
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Cemile Basgul
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Daniel W MacDonald
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Keith Fitzgerald
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven M Kurtz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA; Exponent, Inc., Philadelphia, Pennsylvania, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
64
|
Shivaee A, Sadeghi Kalani B, Talebi M, Darban-Sarokhalil D. Does biofilm formation have different pathways in Staphylococcus aureus?. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 22:1147-1152. [PMID: 31998455 PMCID: PMC6885392 DOI: 10.22038/ijbms.2019.34888.8281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Objective(s): Biofilm formation is one of the most important factors in the development of infections caused by Staphylococcus aureus. In this study, the expression levels of genes responsible for biofilm formation were studied in methicillin sensitive and methicillin resistant S. aureus. Materials and Methods: A total of 100 meticillin-resistant s.aureus (MRSA) and meticillin-sensetive s.aureus (MSSA) isolates were studied. Bacterial biofilm formation was evaluated phenotypically using microtiter plate method. Real-time PCR tests were conducted to determine the expression levels of genes involved in biofilm formation. Results: Quantitative biofilm formation test was repeated three times for each specimen. The prevalence of weak, medium, and strong biofilm producers were 16%, 49%, and 35%, respectively. In MSSA isolates, expression levels of ica genes increased compared to the fnbA, fnbB, clfA and clfB genes. These results were different in MRSA isolates, and ica genes showed a decreased gene expression levels compared to the aforementioned genes. Conclusion: Considering the results of this study, clf genes probably contribute to the same extent in both MRSA and MSSA isolates, and there is probably no significant difference in the role of these genes in these isolates. In addition, the results of this study indicated that MRSA may not use the conventional route for biofilm formation and may use independent pathways through Polysaccharide intercellular adhesion (PIA).
Collapse
Affiliation(s)
- Ali Shivaee
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behrooz Sadeghi Kalani
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
65
|
Macias-Valcayo A, Staats A, Aguilera-Correa JJ, Brooks J, Gupta T, Dusane D, Stoodley P, Esteban J. Synovial Fluid Mediated Aggregation of Clinical Strains of Four Enterobacterial Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1323:81-90. [PMID: 32797406 DOI: 10.1007/5584_2020_573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Septic arthritis and prosthetic joint infection (PJI) are conditions commonly associated with Gram-positive cocci, however, a drastic increase in cases derived from enterobacterial species has been observed. Recently it has been reported by multiple groups that staphylococci rapidly form free-floating aggregates in the presence of synovial fluid. These aggregates are comparatively more resistant to antimicrobial challenge than their planktonic counterparts, and thus may play a role in the pathogenesis of joint infection. While staphylococcal aggregates have been the primary focus of interest in the field, it is unclear just how widespread synovial fluid mediated aggregation (SFMA) is in Gram negative enterobacteria (GNE). Through this work we have evaluated SFMA in clinical GNE isolated from PJIs. Two PJI clinical strains each of Enterobacter cloacae, Escherichia coli, Klebsiella pneumonia and Proteus mirabilis strains representing a range of antibiotic susceptibilities were exposed to 10% bovine synovial fluid supernatant (BSF) using a relatively simple, quick semi-quantitative method using an imaging plate reader. BSF stimulated aggregation within 0.5 h both strains of E. cloacae and P. mirabilis and one strain of E.coli. In one strain of P. mirabilis and E.coli, the size of the aggregates significantly increased from 0.5 to 2 h exposure. In contrast, neither K. pneumoniae strain aggregated in BSF. These preliminary findings show that aggregation can occur quickly in GNE, but the extent appears strain and species specific. Further work is required to assess the impact of SFMA on antibiotic tolerance, host innate immunity and the establishment of biofilms.
Collapse
Affiliation(s)
| | - Amelia Staats
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.,Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | | | - Jack Brooks
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Tripti Gupta
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Devendra Dusane
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA. .,Department of Orthopaedics, The Ohio State University, Columbus, OH, USA. .,National Centre for Advanced Tribology at Southampton (nCATS), National Biofilm Innovation Centre (NBIC), Department Mechanical Engineering, University of Southampton, Southampton, UK.
| | | |
Collapse
|
66
|
Cai YM, Chatelet DS, Howlin RP, Wang ZZ, Webb JS. A novel application of Gini coefficient for the quantitative measurement of bacterial aggregation. Sci Rep 2019; 9:19002. [PMID: 31831832 PMCID: PMC6908595 DOI: 10.1038/s41598-019-55567-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/28/2019] [Indexed: 11/30/2022] Open
Abstract
Non-surface attached bacterial aggregates are frequently found in clinical settings associated with chronic infections. Current methods quantifying the extent to which a suspended bacterial population is aggregated mainly rely on: (1) cell size distribution curves that are difficult to be compared numerically among large-scale samples; (2) the average size/proportion of aggregates in a population that do not specify the aggregation patterns. Here we introduce a novel application of Gini coefficient, herein named Aggregation Coefficient (AC), to quantify the aggregation levels of cystic fibrosis Pseudomonas aeruginosa (CF-PA) isolates in vitro using 3D micrographs, Fiji and MATLAB. Different aggregation patterns of five strains were compared statistically using the numerical AC indexes, which correlated well with the size distribution curves plotted by different biovolumes of aggregates. To test the sensitivity of AC, aggregates of the same strains were treated with nitric oxide (NO), a dispersal agent that reduces the biomass of surface attached biofilms. Strains unresponsive to NO were reflected by comparable AC indexes, while those undergoing dispersal showed a significant reduction in AC index, mirroring the changes in average aggregate sizes and proportions. Therefore, AC provides simpler and more descriptive numerical outputs for measuring different aggregation patterns compared to current approaches.
Collapse
Affiliation(s)
- Yu-Ming Cai
- National Biofilms Innovation Centre, Institute of Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - David S Chatelet
- Biomedical Imaging Unit, University of Southampton, Southampton, SO16 6YD, UK
| | - Robert P Howlin
- NIHR Southampton Respiratory Biomedical Research Centre, Southampton, SO16 6YD, UK
| | - Zhi-Zhong Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Minhang District, Shanghai, China
| | - Jeremy S Webb
- National Biofilms Innovation Centre, Institute of Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
67
|
Direct Microscopic Observation of Human Neutrophil-Staphylococcus aureus Interaction In Vitro Suggests a Potential Mechanism for Initiation of Biofilm Infection on an Implanted Medical Device. Infect Immun 2019; 87:IAI.00745-19. [PMID: 31548325 DOI: 10.1128/iai.00745-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 01/29/2023] Open
Abstract
The ability of human neutrophils to clear newly attached Staphylococcus aureus bacteria from a serum-coated glass surface was examined in vitro using time-lapse confocal scanning laser microscopy. Quantitative image analysis was used to measure the temporal change in bacterial biomass, neutrophil motility, and fraction of the surface area policed by neutrophils. In control experiments in which the surface was inoculated with bacteria but no neutrophils were added, prolific bacterial growth was observed. Neutrophils were able to control bacterial growth but only consistently when the neutrophil/bacterium number ratio exceeded approximately 1. When preattached bacteria were given a head start and allowed to grow for 3 h prior to neutrophil addition, neutrophils were unable to maintain control of the nascent biofilm. In these head-start experiments, aggregates of bacterial biofilm with areas of 50 μm2 or larger formed, and the growth of such aggregates continued even when multiple neutrophils attacked a cluster. These results suggest a model for the initiation of a biofilm infection in which a delay in neutrophil recruitment to an abiotic surface allows surface-attached bacteria time to grow and form aggregates that become protected from neutrophil clearance. Results from a computational model of the neutrophil-biofilm surface contest supported this conceptual model and highlighted the stochastic nature of the interaction. Additionally, we observed that both neutrophil motility and clearance of bacteria were impaired when oxygen tension was reduced to 0% or 2% O2.
Collapse
|
68
|
Shivaee A, Sadeghi Kalani B, Talebi M, Darban-Sarokhalil D. Does biofilm formation have different pathways in Staphylococcus aureus?. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019. [PMID: 31998455 DOI: 10.22038/ijbms2019348888281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
OBJECTIVES Biofilm formation is one of the most important factors in the development of infections caused by Staphylococcus aureus. In this study, the expression levels of genes responsible for biofilm formation were studied in methicillin sensitive and methicillin resistant S. aureus. MATERIALS AND METHODS A total of 100 meticillin-resistant s.aureus (MRSA) and meticillin-sensetive s.aureus (MSSA) isolates were studied. Bacterial biofilm formation was evaluated phenotypically using microtiter plate method. Real-time PCR tests were conducted to determine the expression levels of genes involved in biofilm formation. RESULTS Quantitative biofilm formation test was repeated three times for each specimen. The prevalence of weak, medium, and strong biofilm producers were 16%, 49%, and 35%, respectively. In MSSA isolates, expression levels of ica genes increased compared to the fnbA, fnbB, clfA and clfB genes. These results were different in MRSA isolates, and ica genes showed a decreased gene expression levels compared to the aforementioned genes. CONCLUSION Considering the results of this study, clf genes probably contribute to the same extent in both MRSA and MSSA isolates, and there is probably no significant difference in the role of these genes in these isolates. In addition, the results of this study indicated that MRSA may not use the conventional route for biofilm formation and may use independent pathways through Polysaccharide intercellular adhesion (PIA).
Collapse
Affiliation(s)
- Ali Shivaee
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behrooz Sadeghi Kalani
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
69
|
Structural Insights into Curli CsgA Cross-β Fibril Architecture Inspire Repurposing of Anti-amyloid Compounds as Anti-biofilm Agents. PLoS Pathog 2019; 15:e1007978. [PMID: 31469892 PMCID: PMC6748439 DOI: 10.1371/journal.ppat.1007978] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 09/17/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022] Open
Abstract
Curli amyloid fibrils secreted by Enterobacteriaceae mediate host cell adhesion and contribute to biofilm formation, thereby promoting bacterial resistance to environmental stressors. Here, we present crystal structures of amyloid-forming segments from the major curli subunit, CsgA, revealing steric zipper fibrils of tightly mated β-sheets, demonstrating a structural link between curli and human pathological amyloids. D-enantiomeric peptides, originally developed to interfere with Alzheimer's disease-associated amyloid-β, inhibited CsgA fibrillation and reduced biofilm formation in Salmonella typhimurium. Moreover, as previously shown, CsgA fibrils cross-seeded fibrillation of amyloid-β, providing support for the proposed structural resemblance and potential for cross-species amyloid interactions. The presented findings provide structural insights into amyloidogenic regions important for curli formation, suggest a novel strategy for disrupting amyloid-structured biofilms, and hypothesize on the formation of self-propagating prion-like species originating from a microbial source that could influence neurodegenerative diseases.
Collapse
|
70
|
Llamazares C, Sanz Del Olmo N, Ortega P, Gómez R, Soliveri J, de la Mata FJ, García-Gallego S, Copa-Patiño JL. Antibacterial Effect of Carbosilane Metallodendrimers in Planktonic Cells of Gram-Positive and Gram-Negative Bacteria and Staphylococcus aureus Biofilm. Biomolecules 2019; 9:biom9090405. [PMID: 31450779 PMCID: PMC6769849 DOI: 10.3390/biom9090405] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/17/2019] [Accepted: 08/21/2019] [Indexed: 02/03/2023] Open
Abstract
Antibiotic resistance is currently one of the main threats to public health security. Biofilm formation is a resistance mechanism that is responsible for most human bacterial infections and requires new and effective therapeutic approaches, such as those provided by nanotechnology. In this work, the antibacterial effect of carbosilane metallodendrimers with different metals (copper(II) and ruthenium(II)), ligands (chloride and nitrate) and generations (generation 0, 1 and 2) has been studied using planktonic Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli) bacteria. Furthermore, the ability of the metallodendrimers to avoid the formation of S. aureus biofilms was also evaluated. The results showed a promising biocide activity in both types of planktonic bacteria, especially for first-generation dendrimers, which arises from the metal complexation to the dendrimer. Cu(II) metallodendrimers require lower concentration than Ru(II) counterpart to inhibit the production of S. aureus biofilms, but none produce hemolysis at the inhibitory concentrations and can be safely used as antibacterial agents. In particular, the first-generation Cu(II) metallodendrimer with nitrate ligands displayed the most promising properties to continue with further studies in both planktonic cells and biofilms.
Collapse
Affiliation(s)
- Celia Llamazares
- Department of Biomedicine and Biotechnology, University of Alcalá, 28805 Madrid, Spain
| | - Natalia Sanz Del Olmo
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Paula Ortega
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Juan Soliveri
- Department of Biomedicine and Biotechnology, University of Alcalá, 28805 Madrid, Spain
| | - F Javier de la Mata
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - Sandra García-Gallego
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - José Luis Copa-Patiño
- Department of Biomedicine and Biotechnology, University of Alcalá, 28805 Madrid, Spain
| |
Collapse
|
71
|
Tan J, Wang J, Yang C, Zhu C, Guo G, Tang J, Shen H. Antimicrobial characteristics of Berberine against prosthetic joint infection-related Staphylococcus aureus of different multi-locus sequence types. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:218. [PMID: 31419978 PMCID: PMC6697971 DOI: 10.1186/s12906-019-2558-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Staphylococcal aureus (S. aureus) has become the leading causative pathogen of Prosthetic Joint Infection (PJI), which is the most devastating complication after arthroplasty surgeries. Due to the biofilm formation ability and emergence of multiple-drugs resistance strains of S. aureus, it has become an urgency to find new anti-staphylococcal agents to establish effective prophylaxis and treatment strategy for PJI. Extracted from a traditional Chinese herb, berberine is proved active in inhibiting S. aureus, while whether it exerts the same effect on PJI-related S. aureus remains unknown. This study aims to investigate the antimicrobial activity of berbrine against clinical derived PJI-related S. aureus and whether its inhibiting efficacy is associated with subtypes of S. aureus. METHODS Eighteen PJI-associated S. aureus were collected and their Multi-locus Sequence Types (MLST) and susceptibility to berberine both in planktonic and biofilm form were investigated. Additionally, one S. aureus strain (ST1792) was selected from the group and its transcriptomic profiling in berberine incubation was performed. The statistical analyses were conducted using Student's t-test with SPSS 24.0(SPSS, IBM, USA). The data were expressed as the means ± standard deviation. Values of p < 0.05 were considered statistically significant. RESULTS It was found out that the Minimum Inhibitory Concentration values of PJI-related S. aureus varied in a broad range (from 64 to 512 μg/ml) among different MLST subtypes and the bacteria were able to regain growth after 24 h in berberine of MIC value or higher concentrations. In addition, sub-inhibitory concentrations of berberine surprisingly enhanced biofilm formation in some S. aureus strains. CONCLUSION Traditional medicine is utilised by a large number of individuals, which provides abundant resources for modern medical science. In our study, berberine was found bactericidal against PJI related S. aureus, however, its antibacterial property was impacted by the MLST subtypes of the bacteria, both in planktonic and biofilm growth forms.
Collapse
|
72
|
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: 47] [Impact Index Per Article: 9.4] [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
|
73
|
The MSCRAMM Family of Cell-Wall-Anchored Surface Proteins of Gram-Positive Cocci. Trends Microbiol 2019; 27:927-941. [PMID: 31375310 DOI: 10.1016/j.tim.2019.06.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/10/2019] [Accepted: 06/19/2019] [Indexed: 01/21/2023]
Abstract
The microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are a family of proteins that are defined by the presence of two adjacent IgG-like folded subdomains. These promote binding to ligands by mechanisms that involve major conformational changes exemplified by the binding to fibrinogen by the 'dock-lock-latch' mechanism or to collagen by the 'collagen hug'. Clumping factors A and B are two such MSCRAMMs that have several important roles in the pathogenesis of Staphylococcus aureus infections. MSCRAMM architecture, ligand binding, and roles in infection and colonization are examined with a focus on recent developments with clumping factors.
Collapse
|
74
|
Delaney LJ, MacDonald D, Leung J, Fitzgerald K, Sevit AM, Eisenbrey JR, Patel N, Forsberg F, Kepler CK, Fang T, Kurtz SM, Hickok NJ. Ultrasound-triggered antibiotic release from PEEK clips to prevent spinal fusion infection: Initial evaluations. Acta Biomater 2019; 93:12-24. [PMID: 30826477 PMCID: PMC6764442 DOI: 10.1016/j.actbio.2019.02.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 02/21/2019] [Accepted: 02/26/2019] [Indexed: 01/01/2023]
Abstract
Despite aggressive peri-operative antibiotic treatments, up to 10% of patients undergoing instrumented spinal surgery develop an infection. Like most implant-associated infections, spinal infections persist through colonization and biofilm formation on spinal instrumentation, which can include metal screws and rods for fixation and an intervertebral cage commonly comprised of polyether ether ketone (PEEK). We have designed a PEEK antibiotic reservoir that would clip to the metal fixation rod and that would achieve slow antibiotic release over several days, followed by a bolus release of antibiotics triggered by ultrasound (US) rupture of a reservoir membrane. We have found using human physiological fluid (synovial fluid), that higher levels (100–500 μg) of vancomycin are required to achieve a marked reduction in adherent bacteria vs. that seen in the common bacterial medium, trypticase soy broth. To achieve these levels of release, we applied a polylactic acid coating to a porous PEEK puck, which exhibited both slow and US-triggered release. This design was further refined to a one-hole or two-hole cylindrical PEEK reservoir that can clip onto a spinal rod for clinical use. Short-term release of high levels of antibiotic (340 ± 168 μg), followed by US-triggered release was measured (7420 ± 2992 μg at 48 h). These levels are sufficient to prevent adhesion of Staphylococcus aureus to implant materials. This study demonstrates the feasibility of an US-mediated antibiotic delivery device, which could be a potent weapon against spinal surgical site infection.
Collapse
Affiliation(s)
- Lauren J Delaney
- Department of Radiology, Thomas Jefferson University, 132 S. 10th Street, Philadelphia, PA 19107, USA
| | - Daniel MacDonald
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | - Jay Leung
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | - Keith Fitzgerald
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA
| | - Alex M Sevit
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, 132 S. 10th Street, Philadelphia, PA 19107, USA
| | - Neil Patel
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, 132 S. 10th Street, Philadelphia, PA 19107, USA
| | - Christopher K Kepler
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA; The Rothman Institute, Thomas Jefferson University, 925 Chestnut Street, Philadelphia, PA 19107, USA
| | - Taolin Fang
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA; The Rothman Institute, Thomas Jefferson University, 925 Chestnut Street, Philadelphia, PA 19107, USA
| | - Steven M Kurtz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA; Exponent, Inc., 3440 Market Street Suite 600, Philadelphia, PA 19104, USA
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
75
|
Foster TJ. Surface Proteins of Staphylococcus aureus. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0046-2018. [PMID: 31267926 PMCID: PMC10957221 DOI: 10.1128/microbiolspec.gpp3-0046-2018] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The surface of Staphylococcus aureus is decorated with over 20 proteins that are covalently anchored to peptidoglycan by the action of sortase A. These cell wall-anchored (CWA) proteins can be classified into several structural and functional groups. The largest is the MSCRAMM family, which is characterized by tandemly repeated IgG-like folded domains that bind peptide ligands by the dock lock latch mechanism or the collagen triple helix by the collagen hug. Several CWA proteins comprise modules that have different functions, and some individual domains can bind different ligands, sometimes by different mechanisms. For example, the N-terminus of the fibronectin binding proteins comprises an MSCRAMM domain which binds several ligands, while the C-terminus is composed of tandem fibronectin binding repeats. Surface proteins promote adhesion to host cells and tissue, including components of the extracellular matrix, contribute to biofilm formation by stimulating attachment to the host or indwelling medical devices followed by cell-cell accumulation via homophilic interactions between proteins on neighboring cells, help bacteria evade host innate immune responses, participate in iron acquisition from host hemoglobin, and trigger invasion of bacteria into cells that are not normally phagocytic. The study of genetically manipulated strains using animal infection models has shown that many CWA proteins contribute to pathogenesis. Fragments of CWA proteins have the potential to be used in multicomponent vaccines to prevent S. aureus infections.
Collapse
|
76
|
Abstract
Implant-related infection is one of the leading reasons for failure in orthopaedics and trauma, and results in high social and economic costs. Various antibacterial coating technologies have proven to be safe and effective both in preclinical and clinical studies, with post-surgical implant-related infections reduced by 90% in some cases, depending on the type of coating and experimental setup used. Economic assessment may enable the cost-to-benefit profile of any given antibacterial coating to be defined, based on the expected infection rate with and without the coating, the cost of the infection management, and the cost of the coating. After reviewing the latest evidence on the available antibacterial coatings, we quantified the impact caused by delaying their large-scale application. Considering only joint arthroplasties, our calculations indicated that for an antibacterial coating, with a final user's cost price of €600 and able to reduce post-surgical infection by 80%, each year of delay to its large-scale application would cause an estimated 35 200 new cases of post-surgical infection in Europe, equating to additional hospital costs of approximately €440 million per year. An adequate reimbursement policy for antibacterial coatings may benefit patients, healthcare systems, and related research, as could faster and more affordable regulatory pathways for the technologies still in the pipeline. This could significantly reduce the social and economic burden of implant-related infections in orthopaedics and trauma. Cite this article: C. L. Romanò, H. Tsuchiya, I. Morelli, A. G. Battaglia, L. Drago. Antibacterial coating of implants: are we missing something? Bone Joint Res 2019;8:199-206. DOI: 10.1302/2046-3758.85.BJR-2018-0316.
Collapse
Affiliation(s)
- C. L. Romanò
- Studio Medico Associato Cecca-Romanò, Milan, Italy
| | - H. Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - I. Morelli
- Specialty School of Orthopaedics, University of Milan, Milan, Italy
| | - A. G. Battaglia
- Specialty School of Orthopaedics, University of Milan, Milan, Italy
| | - L. Drago
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
77
|
Chen C, Yan Q, Tao M, Shi H, Han X, Jia L, Huang Y, Zhao L, Wang C, Ma X, Ma Y. Characterization of serine acetyltransferase (CysE) from methicillin-resistant Staphylococcus aureus and inhibitory effect of two natural products on CysE. Microb Pathog 2019; 131:218-226. [PMID: 30974158 DOI: 10.1016/j.micpath.2019.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 01/19/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major hospital-acquired infective pathogen that has developed resistance to many antibiotics. It is imperious to develop novel anti-MRSA drugs to control the emergence of drug resistance. The biosynthesis of cysteine in bacteria is catalyzed by CysE and CysK. CysE was predicted to be important for bacterial viability, it could be a potential drug target. The serine acetyltransferase activity of CysE was detected and its catalytic properties were also determined. CysE homology model was built to investigate interaction sites between CysE and substrate L-Ser or inhibitors by molecular docking. Docking data showed that residues Asp94 and His95 were essential for serine acetyltransferase activity of CysE, which were confirmed by site-directed mutagenesis. Colorimetric assay was used to screen natural products and six compounds which inhibited CysE activity (IC50 ranging from 29.83 μM to 203.13 μM) were found. Inhibition types of two compounds 4 (11-oxo-ebracteolatanolide B) and 30 ((4R,4aR)-dihydroxy-3-hydroxymethyl-7,7,10a-trimethyl-2,4,4a,5,6,6a,7,8,9,10,10a,l0b-dodecahydrophenanthro[3,2-b]furan-2-one) on CysE were determined. Compounds 4 and 30 showed inhibitory effect on MRSA growth (MIC at 12.5 μg/ml and 25 μg/ml) and mature biofilm. The established colorimetric assay will facilitate further high-throughput screening of CysE inhibitors from different compound libraries. The compounds 4 and 30 may offer structural basis for developing new anti-MRSA drugs.
Collapse
Affiliation(s)
- Changming Chen
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Qiulong Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Mengxing Tao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Huaying Shi
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Xiuyan Han
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Liqiu Jia
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Yukun Huang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Lizhe Zhao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Chao Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xiaochi Ma
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yufang Ma
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
78
|
Wichmann T, Moriarty TF, Keller I, Pfister S, Deggim-Messmer V, Gautier E, Kalberer F, Koch PP, Wahl P. Prevalence and quantification of contamination of knitted cotton outer gloves during hip and knee arthroplasty surgery. Arch Orthop Trauma Surg 2019; 139:451-459. [PMID: 30406429 DOI: 10.1007/s00402-018-3061-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Knitted cotton outer gloves offer protection against surgical glove perforation and provide improved grip on instruments. These gloves absorb blood and other fluids during surgery, and may therefore also accumulate contaminating bacteria. To date, there is no published data on microbial contamination of such gloves during surgery. METHODS Knitted cotton outer gloves used in primary and revision hip and knee arthroplasty from two Swiss hospitals were analysed by quantitative bacteriology. Samples were subjected to sonication and vortexing, followed by membrane filtration of the sonicate. Membranes were incubated under aerobic and anaerobic culture conditions, respectively, for 21 days. Total microbial load for each pair of gloves was determined by colony-forming units (CFU) count. Strain identification was performed with MALDI-TOF. RESULTS A total of 43 pairs of gloves were collected from continuous series of surgeries. Under aerobic culture conditions, total CFU counts ranged 0-1103, 25 (58%) samples remaining sterile, and 4 (9%) yielding > 100 CFU. Under anaerobic culture conditions, total CFU counts ranged 0-3579, 22 (51%) samples remaining sterile, 6 (14%) yielding > 100 CFU. The only covariate significantly associated with the level of contamination was the provider hospital (p < 0.0001 for aerobic and p = 0.007 for anaerobic cultures). Strain identification revealed only skin commensals, mainly coagulase-negative staphylococci and Propionibacterium spp. CONCLUSION While contamination of surgical latex gloves is a well-known issue, no study has examined so far contamination of knitted cotton outer gloves. No or very low microbial contamination could be identified in the majority of the knitted cotton outer gloves assayed. However, a relevant proportion showed contamination far higher than estimated minimal thresholds for implant-associated infection. Clinical relevance of these findings remains to be established.
Collapse
Affiliation(s)
- Thorsten Wichmann
- School for Medical Technology and Medical Computer Sciences, Bernese University of Applied Sciences, Bern, Switzerland.,Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Brauerstrasse 15, P.O. Box 834, 8401, Winterthur, Switzerland
| | | | - Iris Keller
- AO Research Institute Davos, Davos, Switzerland
| | - Stefan Pfister
- Microbiology Laboratories, HFR Fribourg-Cantonal Hospital, Fribourg, Switzerland
| | | | - Emanuel Gautier
- Department for Orthopaedic Surgery, HFR Fribourg-Cantonal Hospital, Fribourg, Switzerland
| | - Fabian Kalberer
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Brauerstrasse 15, P.O. Box 834, 8401, Winterthur, Switzerland
| | - Peter P Koch
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Brauerstrasse 15, P.O. Box 834, 8401, Winterthur, Switzerland
| | - Peter Wahl
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Brauerstrasse 15, P.O. Box 834, 8401, Winterthur, Switzerland.
| |
Collapse
|
79
|
Luo Z, Chen M, Chen T, She P, Wu Y. Lactic Acid Produced by Glycolysis Contributed to Staphylococcus aureus Aggregation Induced by Glucose. Curr Microbiol 2019; 76:607-612. [PMID: 30895345 DOI: 10.1007/s00284-019-01666-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/01/2019] [Indexed: 10/27/2022]
Abstract
High concentration of glucose induces Staphylococcus aureus (S. aureus) aggregation, but the mechanism of this is still unclear. In this study, the aggregation of S. aureus strains was induced by high concentration of glucose (>7.8 mM), and which was dose- and time-dependent. In addition, the large amount of lactate acid produced during S. aureus aggregation, induced by glucose, resulted in decreased pH value. Lactic acid, the end product of glycolysis, could quickly induce S. aureus aggregation. Except for lactic acid, acetic acid and HCl also induced S. aureus aggregation. In addition, the aggregation of S. aureus strains induced by glucose or lactic acid was completely inhibited in Tris-HCl buffer (pH 7.5), and inhibition of glycolysis by 2-deoxyglucose significantly decreased S. aureus aggregation. The aggregation induced by glucose was dispersed by periodate and proteinase K. In summary, lactate acid produced by glycolysis contributed to S. aureus aggregation induced by high concentration of glucose.
Collapse
Affiliation(s)
- Zhen Luo
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Meilin Chen
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ti Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
80
|
Bastos LR, Martins MCF, Albano RM, Marques EA, Leão RS. Whole genome sequencing of a ST2594 MRSA strain causing non-mucosal preoperative colonization and low-grade postoperative infection. Antonie van Leeuwenhoek 2019; 112:961-964. [PMID: 30663019 DOI: 10.1007/s10482-019-01229-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 01/09/2019] [Indexed: 10/27/2022]
Abstract
We present a post-operative infection caused by a methicillin-resistant Staphylococcus aureus strain, previously isolated in the preoperative screening, in a patient submitted to femoral osteosynthesis, successfully treated with oral ciprofloxacin. The isolate exhibited in vitro resistance to ciprofloxacin, Staphylococcal Cassette Chromosome mec type IV, it was negative for the lukS-PV Panton-Valentine leucocidin gene and belonged to ST2594 in multilocus sequence typing analysis. Whole genome sequencing revealed a genome size of 2,818,289 base pairs. The annotated genomes of ST2594 and N315 strains were compared, looking for genes related to virulence and resistance. The lack of the tst, sec, sel genes, associated with a mutation in the clfA gene, may partially explain the low morbity in this case.
Collapse
Affiliation(s)
- Leonardo Ribeiro Bastos
- Department of Microbiology, Immunology and Parasitology, Medical Science School, State University of Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, 3º andar, Rio de Janeiro, RJ, CEP: 20551-030, Brazil
| | | | - Rodolpho Mattos Albano
- Biochemistry Department, Institute of Biology Roberto Alcântara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elizabeth Andrade Marques
- Department of Microbiology, Immunology and Parasitology, Medical Science School, State University of Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, 3º andar, Rio de Janeiro, RJ, CEP: 20551-030, Brazil
| | - Robson Souza Leão
- Department of Microbiology, Immunology and Parasitology, Medical Science School, State University of Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, 3º andar, Rio de Janeiro, RJ, CEP: 20551-030, Brazil.
| |
Collapse
|
81
|
Raafat D, Otto M, Reppschläger K, Iqbal J, Holtfreter S. Fighting Staphylococcus aureus Biofilms with Monoclonal Antibodies. Trends Microbiol 2019; 27:303-322. [PMID: 30665698 DOI: 10.1016/j.tim.2018.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a notorious pathogen and one of the most frequent causes of biofilm-related infections. The treatment of S. aureus biofilms is hampered by the ability of the biofilm structure to shield bacteria from antibiotics as well as the host's immune system. Therefore, new preventive and/or therapeutic interventions, including the use of antibody-based approaches, are urgently required. In this review, we describe the mechanisms by which anti-S. aureus antibodies can help in combating biofilms, including an up-to-date overview of monoclonal antibodies currently in clinical trials. Moreover, we highlight ongoing efforts in passive vaccination against S. aureus biofilm infections, with special emphasis on promising targets, and finally indicate the direction into which future research could be heading.
Collapse
Affiliation(s)
- Dina Raafat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Egypt; Current affiliation: Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Kevin Reppschläger
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jawad Iqbal
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
82
|
Abstract
Staphylococci, with the leading species Staphylococcus aureus and Staphylococcus epidermidis, are the most frequent causes of infections on indwelling medical devices. The biofilm phenotype that those bacteria adopt during device-associated infection facilitates increased resistance to antibiotics and host immune defenses. This review presents and discusses the molecular mechanisms contributing to staphylococcal biofilm development and their in-vivo importance. Furthermore, it summarizes current strategies for the development of therapeutics against staphylococcal biofilm-associated infection.
Collapse
|
83
|
Badha V, Moore R, Heffernan J, Castaneda P, McLaren A, Overstreet D. Determination of Tobramycin and Vancomycin Exposure Required to Eradicate Biofilms on Muscle and Bone Tissue In Vitro. J Bone Jt Infect 2019; 4:1-9. [PMID: 30755841 PMCID: PMC6367195 DOI: 10.7150/jbji.29711] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/10/2018] [Indexed: 01/09/2023] Open
Abstract
Background: Bacterial biofilms cause chronic orthopaedic infections. Surgical debridement to remove biofilm can be ineffective without adjuvant local antimicrobials because undetected biofilm fragments may remain in the wound and reestablish the infection if untreated. However, the concentrations and duration of antimicrobial exposure necessary to eradicate bacteria from clinical biofilms remain largely undefined. In this study, we determined the minimum biofilm eradication concentration (MBEC) of tobramycin and vancomycin for bacterial biofilms grown on bone and muscle in vitro. Methods: Biofilms of pathogens found in musculoskeletal infections (S. aureus, S. epidermidis, E. faecalis, P. aeruginosa, and E. coli) were established for 72 hr on rabbit muscle and bone specimens in vitro and characterized by SEM imaging and CFU counts. Biofilm-covered tissue specimens were exposed to serial log2 dilutions (4000-31.25 µg/mL) of tobramycin, vancomycin, or a 1:1 combination of both drugs for 6, 24, or 72 hr. Tissues were subcultured following antimicrobial exposure to determine bacterial survival. The breakpoint concentration with no surviving bacteria was defined as the MBEC for each pathogen-antimicrobial-exposure time combination. Results: All tested pathogens formed biofilm on tissue. Tobramycin/vancomycin (1:1) was the most effective antimicrobial regimen with MBEC on muscle (10/10 pathogens) or bone (7/10 pathogens) generally in the range of 100-750 µg/mL with 24 or 72 hr exposure. MBEC decreased with exposure time for 53.3% of biofilms between 6 and 24 hr, 53.3% of biofilms between 24 and 72 hr, and for 76.7% of biofilms between 6 and 72 hr. MBECs on bone were significantly higher than corresponding MBECs on muscle tissue (p < 0.05). In most cases, tissue MBECs were lower compared to previously published MBECs for the same pathogens on polystyrene tissue-culture plates. Conclusions: The majority of MBECs for orthopaedic infections on bone and muscle are on the order of 100-750 µg/mL of vancomycin+tobramycin when sustained for at least 24 hr, which may be clinically achievable using high-dose antimicrobial-loaded bone cement (ALBC).
Collapse
Affiliation(s)
- Vajra Badha
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
| | - Rex Moore
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
| | | | | | - Alex McLaren
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
- University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Derek Overstreet
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
| |
Collapse
|
84
|
Krogh AKH, Haaber J, Bochsen L, Ingmer H, Kristensen AT. Aggregating resistant Staphylococcus aureus induces hypocoagulability, hyperfibrinolysis, phagocytosis, and neutrophil, monocyte, and lymphocyte binding in canine whole blood. Vet Clin Pathol 2018; 47:560-574. [PMID: 30586190 DOI: 10.1111/vcp.12679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 06/07/2018] [Accepted: 06/24/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Staphylococcus aureus is an opportunistic pathogen with the ability to form mobile planktonic aggregates during growth, in vitro. The in vivo pathophysiologic effects of S aureus aggregates on host responses are unknown. Knowledge of these could aid in combating infections. OBJECTIVE This study aimed to investigate the effect of increasing concentrations of two different aggregating S aureus strains on the hemostatic and inflammatory host responses in canine whole blood. The hypothesis was that aggregating bacteria would induce pronounced hemostatic and inflammatory responses. METHODS Citrate-stabilized whole blood from 10 healthy dogs was incubated with two strains of aggregating S aureus at three different concentrations. Each sample was analyzed using tissue factor-thromboelastography (TF-TEG) and the formed clot was investigated with electron microscopy. The plasma activated partial thromboplastin time (aPTT), prothrombin time (PT), fibrinogen, and D-dimer tests were measured. Bacteria-leukocyte binding was evaluated with flow cytometry, and neutrophil phagocytosis was assessed using light and transmission electron microscopy. RESULTS The highest concentration of bacteria resulted in a significantly shortened TF-TEG initiation time, decreased alpha, maximum amplitude, global strength, and increased lysis. In addition, significantly shortened PT, decreased fibrinogen, and increased D-dimers were demonstrated at the highest concentration of bacteria. Lower concentrations of bacteria showed no differences in TF-TEG when compared with controls. The findings were similar for both S aureus strains. Increased concentration-dependent binding of bacteria and leukocytes and neutrophil bacterial phagocytosis was observed. CONCLUSIONS Two strains of S aureus induced alterations of clot formation in concentrations where bacterial aggregates were formed. A concentration-dependent cellular inflammatory response was observed.
Collapse
Affiliation(s)
- Anne K H Krogh
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jakob Haaber
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Louise Bochsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Annemarie T Kristensen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
85
|
Baghini GS, Sepahi AA, Tabatabaei RR, Tahvildari K. The combined effects of ethanolic extract of Artemisia aucheri and Artemisia oliveriana on biofilm genes expression of methicillin resistant Staphylococcus aureus. IRANIAN JOURNAL OF MICROBIOLOGY 2018; 10:417-423. [PMID: 30873270 PMCID: PMC6414741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AND OBJECTIVES One of the most important antibiotic-resistant bacteria is methicillin-resistant Staphylococcus aureus (MRSA) biofilm that has caused significant problems in treating the patients. Therefore, the aim of this study was to evaluate the levels of expression of genes involved in biofilm formation in MRSA (ATCC 33591) while being treated by a combination of Artemisia aucheri and Artemisia oliveriana. MATERIALS AND METHODS The minimum inhibitory concentration (MIC) of ethanolic extract of A. aucheri and A. oliveriana and also the minimum inhibitory concentration of combination of both extracts were 512, 1024 and 256 μg/ml, respectively; then at concentrations lower than the MIC, expression levels of the desired genes were determined by Real Time PCR. RESULTS Based on results, using a combination of two ethanolic extracts had a significant effect on expression of genes involved in biofilm formation in MRSA. The expression level of icaA at 4, 8, 16 h after being treated by herbal extracts of A. aucheri and A. oliveriana was 0.293, 0.121, 0.044, respectively. The expression level of icaD was 0.285, 0.097, 0.088, respectively, while that of ebps was 0.087, 0.042, 0.009 at 4, 8 and 16 h, respectively. CONCLUSION This study provided evidence that ethanol extract of A. oliveriava and A. aucheri can inhibit the biofilm formation of S. aureus. As a traditional Iranian medicine, A. oliveriava and A. aucheri extracts have a potential antibiofilm formation against MRSA strains.
Collapse
Affiliation(s)
- Ghazaleh Shojaei Baghini
- Department of Microbiology, Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Abbas Akhavan Sepahi
- Department of Microbiology, Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran,Corresponding author: Abbas Akhavan Sepahi, PhD, Department of Microbiology, Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran. Tel: +98-21-22949793,
| | - Robab Rafiei Tabatabaei
- Department of Microbiology, Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kambiz Tahvildari
- Department of Chemistry, North Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
86
|
Gilbertie JM, Schnabel LV, Stefanovski D, Kelly DJ, Jacob ME, Schaer TP. Gram-negative multi-drug resistant bacteria influence survival to discharge for horses with septic synovial structures: 206 Cases (2010-2015). Vet Microbiol 2018; 226:64-73. [PMID: 30389045 DOI: 10.1016/j.vetmic.2018.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/26/2018] [Accepted: 10/10/2018] [Indexed: 11/29/2022]
Abstract
Bacterial colonization of synovial structures can cause infections that are difficult to treat. Systemic and local antimicrobials and repeated joint lavages are the mainstays of therapy. However, despite aggressive treatments, infection may persist, leading to significant tissue damage or death of the patient. In order to investigate the impact of bacterial culture and antimicrobial resistance on survival to discharge, we reviewed medical records of horses admitted to the University of Pennsylvania's large animal teaching hospital from 2010-2015. Two-hundred and six cases with a definitive diagnosis of septic synovitis and a synovial fluid sample submitted for microbiological culture were included in the study. Of these horses, 48% were culture negative and 52% were positive for any bacterial growth, of which 66% were gram-positive and 28% were gram-negative aerobic organisms with 4% anaerobic and 2% fungal organisms. Overall survival to discharge from hospital was 86%. Horses that had negative growth on culture were more likely to survive until discharge (p < 0.02). Multivariable analyses revealed that the likelihood of euthanasia was significantly associated with identification of coagulase positive Staphylococcus spp. (OR 7.66, 5.46-10.74, p < 0.0001), β-hemolytic Streptococcus spp. (OR 5.18, 3.56-7.55, p < 0.0001), Enterococcus spp. (OR 18.38, 11.45-29.52, p = 0.002), Enterobacteriaceae (OR 31.37, 22.28-44.17, p < 0.0001), Pseudomonas aeruginosa (OR 9.31, 5.30-16.34, p = 0.0004) or other gram-negative species (OR 3.51, 2.07-5.94, p = 0.001). Multi-drug resistance and gram-negative bacteria species were associated with significantly decreased survival rates (OR 119.24, 70.57-201.46, p < 0.0001). In conclusion, prognosis for survival to discharge was poor for horses that were infected with gram-negative organisms, particularly those with MDR phenotypes.
Collapse
Affiliation(s)
- Jessica M Gilbertie
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, 382 West Street Rd., Kennett Square, PA 19348, United States; North Carolina State University, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, United States
| | - Lauren V Schnabel
- North Carolina State University, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, United States
| | - Darko Stefanovski
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, 382 West Street Rd., Kennett Square, PA 19348, United States
| | - Donna J Kelly
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, 382 West Street Rd., Kennett Square, PA 19348, United States
| | - Megan E Jacob
- North Carolina State University, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, United States
| | - Thomas P Schaer
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, 382 West Street Rd., Kennett Square, PA 19348, United States.
| |
Collapse
|
87
|
Jia Z, Zhou W, Yan J, Xiong P, Guo H, Cheng Y, Zheng Y. Constructing Multilayer Silk Protein/Nanosilver Biofunctionalized Hierarchically Structured 3D Printed Ti6Al4 V Scaffold for Repair of Infective Bone Defects. ACS Biomater Sci Eng 2018; 5:244-261. [DOI: 10.1021/acsbiomaterials.8b00857] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Zhaojun Jia
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China
- Center for Biomedical Materials and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Wenhao Zhou
- Center for Biomedical Materials and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jianglong Yan
- Center for Biomedical Materials and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Pan Xiong
- Center for Biomedical Materials and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Hui Guo
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yan Cheng
- Center for Biomedical Materials and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China
- Center for Biomedical Materials and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
88
|
Low-Virulence Organisms and Periprosthetic Joint Infection-Biofilm Considerations of These Organisms. Curr Rev Musculoskelet Med 2018; 11:409-419. [PMID: 29961193 DOI: 10.1007/s12178-018-9503-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The purpose of this manuscript is to provide a critical review of peer-reviewed literature over the last 5 years related to low virulent organisms associated with periprosthetic joint infection (PJI). We evaluated the most common organisms, the diagnostic challenges, and the novel tools available in the perioperative workup of PJI as well as the current understanding of how biofilm potentiates the indolent clinical presentation and explore a possible shift in the surgical management of these patients. RECENT FINDINGS Biofilm actively prevents macrophage phagocytosis by suppressing proinflammatory activity through the recruitment of myeloid-derived suppressor cells. Given the appropriate host and organism conditions, increased utilization of one-stage exchange arthroplasty in the surgical treatment of these low virulent infections may be on the rise. Biomarkers and molecular techniques offer encouraging results to diagnose low virulent organisms and future research focused on the disruption of biofilm may ultimately give rise to improved treatment strategies.
Collapse
|
89
|
Zheng Y, He L, Asiamah TK, Otto M. Colonization of medical devices by staphylococci. Environ Microbiol 2018; 20:3141-3153. [PMID: 29633455 DOI: 10.1111/1462-2920.14129] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/16/2018] [Accepted: 04/03/2018] [Indexed: 12/19/2022]
Abstract
The use of medical devices in modern medicine is constantly increasing. Despite the multiple precautionary strategies that are being employed in hospitals, which include increased hygiene and sterilization measures, bacterial infections on these devices still happen frequently. Staphylococci are among the major causes of medical device infection. This is mostly due to the strong capacity of those bacteria to form device-associated biofilms, which provide resistance to chemical and physical treatments as well as attacks by the host's immune system. Biofilm development is a multistep process with specific factors participating in each step. It is tightly regulated to provide a balance between biofilm expansion and detachment. Detachment from a biofilm on a medical device can lead to severe systemic infection, such as bacteremia and sepsis. While our understanding of staphylococcal biofilm formation has increased significantly and staphylococcal biofilm formation on medical devices is among the best understood biofilm-associated infections, the extensive effort put in preclinical studies with the goal to find novel therapies against staphylococcal device-associated infections has not yet resulted in efficient, applicable therapeutic options for that difficult-to-treat type of disease.
Collapse
Affiliation(s)
- Yue Zheng
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Lei He
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Titus K Asiamah
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
90
|
Cheng Y, Yang H, Yang Y, Huang J, Wu K, Chen Z, Wang X, Lin C, Lai Y. Progress in TiO 2 nanotube coatings for biomedical applications: a review. J Mater Chem B 2018; 6:1862-1886. [PMID: 32254353 DOI: 10.1039/c8tb00149a] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Titanium dioxide nanotubes (TNTs) have drawn wide attention and been extensively applied in the field of biomedicine, due to their large specific surface area, good corrosion resistance, excellent biocompatibility, and enhanced bioactivity. This review describes the preparation of TNTs and the surface modification that entrust the nanotubes with better antibacterial property and enhanced osteoblast adhesion, proliferation, and differentiation. Considering the contact between TNTs' surface and surrounding tissues after implantation, the interactions between TNTs (with properties including their diameter, length, wettability, and crystalline phase) and proteins, platelets, bacteria, and cells are illustrated. The state of the art in the applications of TNTs in dentistry, orthopedic implants, and cardiovascular stents are introduced. In particular, the application of TNTs in biosensing has attracted much attention due to its ability for the rapid diagnosis of diseases. Finally, the difficulties and challenges in the practical application of TNTs are also discussed.
Collapse
Affiliation(s)
- Yan Cheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Hibbitts A, O'Leary C. Emerging Nanomedicine Therapies to Counter the Rise of Methicillin-Resistant Staphylococcus aureus. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E321. [PMID: 29473883 PMCID: PMC5849018 DOI: 10.3390/ma11020321] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/14/2018] [Accepted: 02/19/2018] [Indexed: 12/25/2022]
Abstract
In a recent report, the World Health Organisation (WHO) classified antibiotic resistance as one of the greatest threats to global health, food security, and development. Methicillin-resistant Staphylococcus aureus (MRSA) remains at the core of this threat, with persistent and resilient strains detectable in up to 90% of S. aureus infections. Unfortunately, there is a lack of novel antibiotics reaching the clinic to address the significant morbidity and mortality that MRSA is responsible for. Recently, nanomedicine strategies have emerged as a promising therapy to combat the rise of MRSA. However, these approaches have been wide-ranging in design, with few attempts to compare studies across scientific and clinical disciplines. This review seeks to reconcile this discrepancy in the literature, with specific focus on the mechanisms of MRSA infection and how they can be exploited by bioactive molecules that are delivered by nanomedicines, in addition to utilisation of the nanomaterials themselves as antibacterial agents. Finally, we discuss targeting MRSA biofilms using nano-patterning technologies and comment on future opportunities and challenges for MRSA treatment using nanomedicine.
Collapse
Affiliation(s)
- Alan Hibbitts
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
- Trinity Centre of Bioengineering, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
- Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland.
| | - Cian O'Leary
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
- Trinity Centre of Bioengineering, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
- Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland.
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
92
|
Hook JL, Islam MN, Parker D, Prince AS, Bhattacharya S, Bhattacharya J. Disruption of staphylococcal aggregation protects against lethal lung injury. J Clin Invest 2018; 128:1074-1086. [PMID: 29431734 DOI: 10.1172/jci95823] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/04/2018] [Indexed: 01/23/2023] Open
Abstract
Infection by Staphylococcus aureus strain USA300 causes tissue injury, multiorgan failure, and high mortality. However, the mechanisms by which the bacteria adhere to, then stabilize on, mucosal surfaces before causing injury remain unclear. We addressed these issues through the first real-time determinations of USA300-alveolar interactions in live lungs. We found that within minutes, inhaled USA300 established stable, self-associated microaggregates in niches at curved, but not at flat, regions of the alveolar wall. The microaggregates released α-hemolysin toxin, causing localized alveolar injury, as indicated by epithelial dye loss, mitochondrial depolarization, and cytosolic Ca2+ increase. Spread of cytosolic Ca2+ through intercellular gap junctions to adjoining, uninfected alveoli caused pulmonary edema. Systemic pretreatment with vancomycin, a USA300-cidal antibiotic, failed to protect mice infected with inhaled WT USA300. However, vancomycin pretreatment markedly abrogated mortality in mice infected with mutant USA300 that lacked the aggregation-promoting factor PhnD. We interpret USA300-induced mortality as having resulted from rapid bacterial aggregation in alveolar niches. These findings indicate, for the first time to our knowledge, that alveolar microanatomy is critical in promoting the aggregation and, hence, in causing USA300-induced alveolar injury. We propose that in addition to antibiotics, strategies for bacterial disaggregation may constitute novel therapy against USA300-induced lung injury.
Collapse
Affiliation(s)
- Jaime L Hook
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mohammad N Islam
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | | | - Sunita Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Pediatrics, and
| | - Jahar Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
93
|
Drago L. CORR Insights®: Is Treatment With Dithiothreitol More Effective Than Sonication for the Diagnosis of Prosthetic Joint Infection? Clin Orthop Relat Res 2018; 476:439-440. [PMID: 29389799 PMCID: PMC6259690 DOI: 10.1007/s11999.0000000000000135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
94
|
Liu T, Chen Y, Bi G, Luo J, Du Z, Kong J, Chen Y. Generation of Methicillin-Resistant Staphylococcus Aureus Biofilm Infection in an Immunosuppressed Rat Model. Med Sci Monit 2017; 23:5803-5811. [PMID: 29213029 PMCID: PMC5730015 DOI: 10.12659/msm.907479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is a common pathogen responsible for many related infections, and immunosuppressed individuals are more susceptible. Its pathogenicity is associated with its virulence factors, resistance to antibiotics, and ability to form biofilm (BF). MRSA-BF infections in immunosuppressed patients pose great difficulties to clinical treatment. Material/Methods The study aimed to establish a model of MRSA-BF infection in rats with cyclophosphamide (CTX)-induced immunosuppression. For this, rats were administered CTX on days 1 and 4. White blood cells (WBC) were counted, then rats were inoculated with a clinical MRSA 17546 (t037) on day 5. Rats were sacrificed on days 6–10 and tissue samples were examined by scanning electron microscopy. Results Using the dose of CTX: 150 (mg/kg) + 100 (mg/kg) is better than the other 2 programs as the survival rates of the immunocompromised rats were higher than in the other 2 immunosuppressive groups. The survival rate was not different between rats in the clean environment and in the SPF environment. However, the survival rate was affected by the sample acquisitions. Importantly, WBC counts started to decline on day 4, and then started to rise on day 9. Moreover, MRSA-BFs were formed earlier in immunosuppressed rats compared to the normal rats, as shown by scanning electron microscopy. Conclusions The study successfully established an immunosuppressed rat model of MRSA-BF infection, which provides methodological and data support for establishment of such animal models and is useful reference for related research. Our results may help further investigation of MRSA-BF infection.
Collapse
Affiliation(s)
- Tangjuan Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yan Chen
- Department of Respiratory Disease, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Guan Bi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jin Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Zhongye Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jinliang Kong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yiqiang Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
95
|
Wang J, Zhou H, Guo G, Tan J, Wang Q, Tang J, Liu W, Shen H, Li J, Zhang X. Enhanced Anti-Infective Efficacy of ZnO Nanoreservoirs through a Combination of Intrinsic Anti-Biofilm Activity and Reinforced Innate Defense. ACS APPLIED MATERIALS & INTERFACES 2017; 9:33609-33623. [PMID: 28884578 DOI: 10.1021/acsami.7b08864] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The increasing prevalence of implant-associated infections (IAIs) imposes a heavy burden on patients and medical providers. Bacterial biofilms are recalcitrant to antiseptic drugs and local immune defense and can attenuate host proinflammatory response to interfere with bacterial clearance. Zinc oxide nanoparticles (ZnO NPs) play a dual role in antibacterial and immunomodulatory activities but compromise the cytocompatibility because of their intracellular uptake. Here, ZnO NPs were immobilized on titanium to form homogeneous nanofilms (from discontinuous to continuous) through magnetron sputtering, and the possible antimicrobial activity and immunomodulatory effect of nano-ZnO films were investigated. Nano-ZnO films were found to prohibit sessile bacteria more than planktonic bacteria in vitro, and the antibacterial effect occurred in a dose-dependent manner. Using a novel mouse soft tissue IAI model, the in vivo results revealed that nano-ZnO films possessed outstanding antimicrobial efficacy, which could not be ascribed solely to the intrinsic anti-infective activity of nano-ZnO films observed in vitro. Macrophages and polymorphonuclear leukocytes (PMNs), two important factors in innate immune response, were cocultured with nano-ZnO and bacteria/lipopolysaccharide in vitro, and the nano-ZnO films could enhance the antimicrobial efficacy of macrophages and PMNs through promoting phagocytosis and secretion of inflammatory cytokines. This study provides insights into the anti-infective activity and mechanism of ZnO and consolidates the theoretical basis for future clinical applications of ZnO.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Huaijuan Zhou
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences , Shanghai 200050, China
| | - Geyong Guo
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Jiaqi Tan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Qiaojie Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Jin Tang
- Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Wei Liu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Hao Shen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| | - Jinhua Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences , Shanghai 200050, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong 999077, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University , Shanghai 200233, China
| |
Collapse
|
96
|
Abstract
The staphylococci comprise a diverse genus of Gram-positive, nonmotile commensal organisms that inhabit the skin and mucous membranes of humans and other mammals. In general, staphylococci are benign members of the natural flora, but many species have the capacity to be opportunistic pathogens, mainly infecting individuals who have medical device implants or are otherwise immunocompromised. Staphylococcus aureus and Staphylococcus epidermidis are major sources of hospital-acquired infections and are the most common causes of surgical site infections and medical device-associated bloodstream infections. The ability of staphylococci to form biofilms in vivo makes them highly resistant to chemotherapeutics and leads to chronic diseases. These biofilm infections include osteomyelitis, endocarditis, medical device infections, and persistence in the cystic fibrosis lung. Here, we provide a comprehensive analysis of our current understanding of staphylococcal biofilm formation, with an emphasis on adhesins and regulation, while also addressing how staphylococcal biofilms interact with the immune system. On the whole, this review will provide a thorough picture of biofilm formation of the staphylococcus genus and how this mode of growth impacts the host.
Collapse
|
97
|
Bacteriophage Lysin CF-301, a Potent Antistaphylococcal Biofilm Agent. Antimicrob Agents Chemother 2017; 61:AAC.02666-16. [PMID: 28461319 DOI: 10.1128/aac.02666-16] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/21/2017] [Indexed: 02/07/2023] Open
Abstract
Biofilms pose a unique therapeutic challenge because of the antibiotic tolerance of constituent bacteria. Treatments for biofilm-based infections represent a major unmet medical need, requiring novel agents to eradicate mature biofilms. Our objective was to evaluate bacteriophage lysin CF-301 as a new agent to target Staphylococcus aureus biofilms. We used minimum biofilm-eradicating concentration (MBEC) assays on 95 S. aureus strains to obtain a 90% MBEC (MBEC90) value of ≤0.25 μg/ml for CF-301. Mature biofilms of coagulase-negative staphylococci, Streptococcus pyogenes, and Streptococcus agalactiae were also sensitive to disruption, with MBEC90 values ranging from 0.25 to 8 μg/ml. The potency of CF-301 was demonstrated against S. aureus biofilms formed on polystyrene, glass, surgical mesh, and catheters. In catheters, CF-301 removed all biofilm within 1 h and killed all released bacteria by 6 h. Mixed-species biofilms, formed by S. aureus and Staphylococcus epidermidis on several surfaces, were removed by CF-301, as were S. aureus biofilms either enriched for small-colony variants (SCVs) or grown in human synovial fluid. The antibacterial activity of CF-301 was further demonstrated against S. aureus persister cells in exponential-phase and stationary-phase populations. Finally, the antibiofilm activity of CF-301 was greatly improved in combinations with the cell wall hydrolase lysostaphin when tested against a range of S. aureus strains. In all, the data show that CF-301 is highly effective at disrupting biofilms and killing biofilm bacteria, and, as such, it may be an efficient new agent for treating staphylococcal infections with a biofilm component.
Collapse
|
98
|
Mouse model of hematogenous implant-related Staphylococcus aureus biofilm infection reveals therapeutic targets. Proc Natl Acad Sci U S A 2017; 114:E5094-E5102. [PMID: 28607050 DOI: 10.1073/pnas.1703427114] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Infection is a major complication of implantable medical devices, which provide a scaffold for biofilm formation, thereby reducing susceptibility to antibiotics and complicating treatment. Hematogenous implant-related infections following bacteremia are particularly problematic because they can occur at any time in a previously stable implant. Herein, we developed a model of hematogenous infection in which an orthopedic titanium implant was surgically placed in the legs of mice followed 3 wk later by an i.v. exposure to Staphylococcus aureus This procedure resulted in a marked propensity for a hematogenous implant-related infection comprised of septic arthritis, osteomyelitis, and biofilm formation on the implants in the surgical legs compared with sham-operated surgical legs without implant placement and with contralateral nonoperated normal legs. Neutralizing human monoclonal antibodies against α-toxin (AT) and clumping factor A (ClfA), especially in combination, inhibited biofilm formation in vitro and the hematogenous implant-related infection in vivo. Our findings suggest that AT and ClfA are pathogenic factors that could be therapeutically targeted against Saureus hematogenous implant-related infections.
Collapse
|
99
|
Freeland RB, Morello SL, DeLombaert M, Rajamanickam V. Influence of intravenous regional limb perfusion with amikacin sulfate on Staphylococcus aureus
bioburden in distal limb wounds in horses. Vet Surg 2017; 46:663-674. [DOI: 10.1111/vsu.12638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/30/2016] [Accepted: 12/13/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Russell B. Freeland
- Department of Surgical Sciences; School of Veterinary Medicine, University of Wisconsin-Madison; Madison Wisconsin
| | - Samantha L. Morello
- Department of Surgical Sciences; School of Veterinary Medicine, University of Wisconsin-Madison; Madison Wisconsin
| | - Melissa DeLombaert
- Department of Surgical Sciences; School of Veterinary Medicine, University of Wisconsin-Madison; Madison Wisconsin
| | - Victora Rajamanickam
- Department of Biostatistics and Medical Informatics; School of Medicine and Public Health, Institute for Clinical and Translational Research, University of Wisconsin-Madison; Madison Wisconsin
| |
Collapse
|
100
|
Wang L, Si W, Xue H, Zhao X. A fibronectin-binding protein (FbpA) of Weissella cibaria inhibits colonization and infection of Staphylococcus aureus in mammary glands. Cell Microbiol 2017; 19. [PMID: 28125161 DOI: 10.1111/cmi.12731] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/17/2022]
Abstract
Staphylococcus aureus (S. aureus) is a frequent cause of infections in both humans and animals. Probiotics are known to inhibit colonization of pathogens on host tissues. However, mechanisms for the inhibition are still elusive due to complex host-microbe and microbe-microbe interactions. Here, we show that reduced abilities of S. aureus to infect mammary glands in the presence of Weissella cibaria (W. cibaria) were correlated with its poor adherence to mammary epithelial cells. Such inhibition by W. cibaria isolates was at least partially attributed to a fibronectin-binding protein (FbpA) on this lactic acid bacterium. Three W. cibaria isolates containing fbpA had higher inhibitory abilities than other three LAB isolates without the gene. The fbpA-deficient mutant of W. cibaria isolate LW1, LW1ΔfbpA, lost the inhibitory activity to reduce the adhesion of S. aureus to mammary epithelial cells and was less able to reduce the colonization of S. aureus in mammary glands. Expression of FbpA to the surface of LW1ΔfbpA reversed its inhibitory activities. Furthermore, addition of purified FbpA inhibited S. aureus biofilm formation. Our results suggest that W. cibaria FbpA hinders S. aureus colonization and infection through interfering with the S. aureus invasion pathway mediated by fibronectin-binding proteins and inhibiting biofilm formation of S. aureus.
Collapse
Affiliation(s)
- Liangliang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Wei Si
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China.,Department of Animal Science, McGill University, Quebec, Canada
| | - Huping Xue
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China
| | - Xin Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China.,Department of Animal Science, McGill University, Quebec, Canada
| |
Collapse
|