51
|
Springer BD, Baddour LM, Lockhart PB, Thornhill MH. Antibiotic Prophylaxis for Prosthetic Joint Patients Undergoing Invasive Dental Procedures: Time for a Rethink? J Arthroplasty 2022; 37:1223-1226. [PMID: 35158002 DOI: 10.1016/j.arth.2022.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND In the United States, it has been common practice to recommend that dentists provide antibiotic prophylaxis (AP) before invasive dental procedures (IDPs) to prevent late periprosthetic joint infections (LPJIs) in patients who have prosthetic arthroplasties despite lack of evidence for a causal relationship between IDP and LPJI and a lack of evidence for AP efficacy. METHODS A recent study quantified the IDP incidence over the 15-month period prior to LPJI hospital admissions in the United Kingdom for which dental records were available. A case-crossover analysis compared IDP incidence in the 3 months before LPJI admission with the preceding 12 months. The English population was used because guidelines do not recommend AP and any relationship between IDPs and LPJI should be fully exposed. RESULTS No significant positive association was identified between IDPs and LPJI. Indeed, the incidence of IDPs was lower in the 3 months before LPJI hospital admission than that in the preceding 12 months. CONCLUSION In the absence of a significant positive association between IDPs and LPJI, there is no rationale to administer AP before IDPs in patients with prosthetic joints, particularly given the cost and inconvenience of AP, the risk of adverse drug reactions, and the potential for unnecessary AP use that promotes antibiotic resistance. These results should reassure orthopedic surgeons and their patients that dental care of patients who have prosthetic joints should focus on maintaining good oral hygiene rather than on recommending AP for IDPs. Moreover, it should also reassure those in other countries where AP is not recommended that such guidance is sufficient.
Collapse
Affiliation(s)
- Bryan D Springer
- OrthoCarolina Hip and Knee Center and Atrium Musculoskeletal Institute, Charlotte, NC
| | - Larry M Baddour
- Division of Infectious Diseases, Departments of Medicine and Cardiovascular Disease, Mayo Clinic College of Medicine, Rochester, MN
| | - Peter B Lockhart
- Department of Oral Medicine, Carolinas Medical Center - Atrium Health, Charlotte, NC
| | - Martin H Thornhill
- Department of Oral Medicine, Carolinas Medical Center - Atrium Health, Charlotte, NC; Unit of Oral & Maxillofacial Medicine Surgery and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| |
Collapse
|
52
|
Liu H, Liu Z, Yang B, Lopez Morales J, Nash MA. Optimal Sacrificial Domains in Mechanical Polyproteins: S. epidermidis Adhesins Are Tuned for Work Dissipation. JACS AU 2022; 2:1417-1427. [PMID: 35783175 PMCID: PMC9241160 DOI: 10.1021/jacsau.2c00121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
The opportunistic pathogen Staphylococcus epidermidis utilizes a multidomain surface adhesin protein to bind host components and adhere to tissues. While it is known that the interaction between the SdrG receptor and its fibrinopeptide target (FgB) is exceptionally mechanostable (∼2 nN), the influence of downstream B domains (B1 and B2) is unclear. Here, we studied the mechanical relationships between folded B domains and the SdrG receptor bound to FgB. We used protein engineering, single-molecule force spectroscopy (SMFS) with an atomic force microscope (AFM), and Monte Carlo simulations to understand how the mechanical properties of folded sacrificial domains, in general, can be optimally tuned to match the stability of a receptor-ligand complex. Analogous to macroscopic suspension systems, sacrificial shock absorber domains should neither be too weak nor too strong to optimally dissipate mechanical energy. We built artificial molecular shock absorber systems based on the nanobody (VHH) scaffold and studied the competition between domain unfolding and receptor unbinding. We quantitatively determined the optimal stability of shock absorbers that maximizes work dissipation on average for a given receptor and found that natural sacrificial domains from pathogenic S. epidermidis and Clostridium perfringens adhesins exhibit stabilities at or near this optimum within a specific range of loading rates. These findings demonstrate how tuning the stability of sacrificial domains in adhesive polyproteins can be used to maximize mechanical work dissipation and serve as an adhesion strategy by bacteria.
Collapse
Affiliation(s)
- Haipei Liu
- Department
of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, 4058 Basel, Switzerland
| | - Zhaowei Liu
- Department
of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, 4058 Basel, Switzerland
| | - Byeongseon Yang
- Department
of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, 4058 Basel, Switzerland
| | - Joanan Lopez Morales
- Department
of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, 4058 Basel, Switzerland
| | - Michael A. Nash
- Department
of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department
of Biosystems Science and Engineering, ETH
Zurich, 4058 Basel, Switzerland
| |
Collapse
|
53
|
Austin PD, Stapleton P, Elia M. Comparative effect of seven prophylactic locks to prevent biofilm biomass and viability in intravenous catheters. J Antimicrob Chemother 2022; 77:2191-2198. [PMID: 35723966 DOI: 10.1093/jac/dkac181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Patients requiring long-term intravenous access are at risk of intraluminal catheter bloodstream infection. 'Prophylactic' locks aim to limit this risk but there is uncertainty regarding the most effective lock. OBJECTIVES To develop a novel technique intended to replicate clinical procedures to compare the effectiveness of various 'prophylactic' locks against biofilm biomass ('biomass') formation and biofilm viability ('viability') of Escherichia coli and Staphylococcus epidermidis in intravenous catheters. METHODS For 10 consecutive days 106 cfu/mL E. coli NCTC 10418 and S. epidermidis ATCC 12228 were separately cultured in single lumen 9.6 French silicone tunnelled and cuffed catheters. These were flushed with 0.9% w/v sodium chloride using a push-pause technique before and after instillation of seven 'prophylactic' locks (water, ethanol, sodium chloride, heparinized sodium chloride, citrate, taurolidine plus citrate, and taurolidine; each in triplicate) for 6 h daily. Intraluminal 'biomass' and 'viability' were quantified using crystal violet staining and flush culture, respectively. RESULTS The reduction of 'biomass' and 'viability' depended on both agent and species. Citrate was least effective against E. coli 'viability' and 'biomass' but most effective against S. epidermidis 'viability', and taurolidine was most effective against E. coli 'biomass' and 'viability' but least effective against S. epidermidis 'viability'. 'Biomass' and 'viability' were significantly correlated in E. coli between (r = 0.997, P < 0.001) and within (r = 0.754, P = 0.001) interventions, but not in S. epidermidis. CONCLUSIONS A novel technique found the effect of 'prophylactic' agents in reducing 'biomass' and 'viability' varied by species. The choice of agent depends on the most likely infecting organism.
Collapse
Affiliation(s)
- Peter David Austin
- UCL School of Pharmacy, University College London, London, UK.,Pharmacy Department, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Paul Stapleton
- UCL School of Pharmacy, University College London, London, UK
| | - Marinos Elia
- Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
54
|
Azara E, Longheu CM, Attene S, Sanna S, Sale M, Addis MF, Tola S. Comparative profiling of agr locus, virulence, and biofilm-production genes of human and ovine non-aureus staphylococci. BMC Vet Res 2022; 18:212. [PMID: 35655210 PMCID: PMC9161600 DOI: 10.1186/s12917-022-03257-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
Background In a collaboration between animal and human health care professionals, we assessed the genetic characteristics shared by non-aureus staphylococci (NAS) infecting humans and dairy ewes to investigate their relatedness in a region concentrating half of the total National sheep stock. We examined by PCR 125 ovine and 70 human NAS for biofilm production, pyrogenic toxins, adhesins, autolysins genes, and accessory gene regulator (agr) locus. The microtiter plate assay (MPA) was used for the phenotypic screening of biofilm production. Ovine NAS included S. epidermidis, S. chromogenes, S. haemolyticus, S. simulans, S. caprae, S. warneri, S. saprophyticus, S. intermedius, and S. muscae. Human NAS included S. haemolyticus, S. epidermidis, S. hominis, S. lugdunensis, S. capitis, S. warneri, S. xylosus, S. pasteuri, and S. saprophyticus subsp. bovis. Results Phenotypically, 41 (32.8%) ovine and 24 (34.3%) human isolates were characterized as biofilm producers. Of the ovine isolates, 12 were classified as biofilm-producing while the remaining 29 as weak biofilm-producing. All 24 human isolates were considered weak biofilm-producing. Few S. epidermidis isolates harbored the icaA/D genes coding for the polysaccharide intercellular adhesin (PIA), while the bhp, aap, and embp genes coding biofilm accumulation proteins were present in both non-producing and biofilm-producing isolates. Fifty-nine sheep NAS (all S. epidermidis, 1 S. chromogenes, and 1 S. haemolyticus) and 27 human NAS (all S. epidermidis and 1 S. warneri) were positive for the agr locus: agr-3se (57.8%) followed by agr-1se (36.8%) predominated in sheep, while agr-1se (65.4%), followed by agr-2se (34.6%) predominated in humans. Concerning virulence genes, 40, 39.2, 47.2%, 52.8, 80 and 43.2% of the sheep isolates carried atlE, aae, sdrF, sdrG, eno and epbS respectively, against 37.1, 42.8, 32.8, 60, 100 and 100% of human isolates. Enterotoxins and tsst were not detected. Conclusions Considerable variation in biofilm formation ability was observed among NAS isolates from ovine and human samples. S. epidermidis was the best biofilm producer with the highest prevalence of adhesin-encoding genes. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03257-w.
Collapse
|
55
|
Phenotypic and Molecular Detection of Slime Producing Staphylococcus Spp. Obtained from Blood Samples of Patients Undergoing Hematopoietic Stem-Cell Transplantation. ACTA MEDICA BULGARICA 2022. [DOI: 10.2478/amb-2022-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Aim: to investigate the slime production in isolates of Staphylococcus spp., associated with bacteremia in patients after hematopoietic stem-cell transplantation (HSCT) and to determine the relationship between the slime production and ica genes carriage, as well as the correlation of ica and methicillin resistance.
Materials and methods: Between 2019 and 2020, twenty-one clinically significant Staphylococcus spp. isolates were obtained from blood cultures of 17 patients after HSCT. The species identification and the susceptibility to cefoxitin were determined by BD Phoenix M50. Two phenotypic tests (Congo red agar, CRA; Christensen’s method, TT) and PCR for icaA and icaD were used to detect slime production. A PCR method was also used to detect the mecA, mecC genes.
Results: In the studied group of 21 isolates (S. epidermidis, n = 12; S. haemolyticus, n = 4; S. hominis, n = 2; S. aureus, n = 3), the phenotypic tests were positive in 13 isolates. Ten isolates (47.6%) were identified as carriers of ica genes (S. epidermidis, n = 9, and S. haemolyticus, n = 1). Five isolates (23.8%) were detected as slime producers by all three methods. The mecA gene was identified in 18 isolates (85.7%). All ica positive isolates were also mecA carriers.
Conclusion: A relatively high proportion of the blood isolates of Staphylococcus spp. were slime producers, associ-ated with ica genes. A combination of both phenotypic and genetic methods should be used to detect alternative routes of slime production. The co-expression of ica and mecA is associ-ated with the occurrence of difficult-to-eradicate isolates.
Collapse
|
56
|
Visnapuu A, Van der Gucht M, Wagemans J, Lavigne R. Deconstructing the Phage-Bacterial Biofilm Interaction as a Basis to Establish New Antibiofilm Strategies. Viruses 2022; 14:v14051057. [PMID: 35632801 PMCID: PMC9145820 DOI: 10.3390/v14051057] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/19/2022] Open
Abstract
The bacterial biofilm constitutes a complex environment that endows the bacterial community within with an ability to cope with biotic and abiotic stresses. Considering the interaction with bacterial viruses, these biofilms contain intrinsic defense mechanisms that protect against phage predation; these mechanisms are driven by physical, structural, and metabolic properties or governed by environment-induced mutations and bacterial diversity. In this regard, horizontal gene transfer can also be a driver of biofilm diversity and some (pro)phages can function as temporary allies in biofilm development. Conversely, as bacterial predators, phages have developed counter mechanisms to overcome the biofilm barrier. We highlight how these natural systems have previously inspired new antibiofilm design strategies, e.g., by utilizing exopolysaccharide degrading enzymes and peptidoglycan hydrolases. Next, we propose new potential approaches including phage-encoded DNases to target extracellular DNA, as well as phage-mediated inhibitors of cellular communication; these examples illustrate the relevance and importance of research aiming to elucidate novel antibiofilm mechanisms contained within the vast set of unknown ORFs from phages.
Collapse
|
57
|
Kaur H, Kaur A, Soni SK, Rishi P. Microbially-derived cocktail of carbohydrases as an anti-biofouling agents: a 'green approach'. BIOFOULING 2022; 38:455-481. [PMID: 35673761 DOI: 10.1080/08927014.2022.2085566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Enzymes, also known as biocatalysts, display vital properties like high substrate specificity, an eco-friendly nature, low energy inputs, and cost-effectiveness. Among their numerous known applications, enzymes that can target biofilms or their components are increasingly being investigated for their anti-biofouling action, particularly in healthcare, food manufacturing units and environmental applications. Enzymes can target biofilms at different levels like during the attachment of microorganisms, formation of exopolymeric substances (EPS), and their disruption thereafter. In this regard, a consortium of carbohydrases that can target heterogeneous polysaccharides present in the EPS matrix may provide an effective alternative to conventional chemical anti-biofouling methods. Further, for complete annihilation of biofilms, enzymes can be used alone or in conjunction with other antimicrobial agents. Enzymes hold the promise to replace the conventional methods with greener, more economical, and more efficient alternatives. The present article explores the potential and future perspectives of using carbohydrases as effective anti-biofilm agents.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Arashdeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
58
|
Low Concentration of the Neutrophil Proteases Cathepsin G, Cathepsin B, Proteinase-3 and Metalloproteinase-9 Induce Biofilm Formation in Non-Biofilm-Forming Staphylococcus epidermidis Isolates. Int J Mol Sci 2022; 23:ijms23094992. [PMID: 35563384 PMCID: PMC9102557 DOI: 10.3390/ijms23094992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/04/2022] Open
Abstract
Neutrophils play a crucial role in eliminating bacteria that invade the human body; however, cathepsin G can induce biofilm formation in a non-biofilm-forming Staphylococcus epidermidis 1457 strain, suggesting that neutrophil proteases may be involved in biofilm formation. Cathepsin G, cathepsin B, proteinase-3, and metalloproteinase-9 (MMP-9) from neutrophils were tested on the biofilm induction in commensal (skin isolated) and clinical non-biofilm-forming S. epidermidis isolates. From 81 isolates, 53 (74%) were aap+, icaA−, icaD− genotype, and without the capacity of biofilm formation under conditions of 1% glucose, 4% ethanol or 4% NaCl, but these 53 non-biofilm-forming isolates induced biofilm by the use of different neutrophil proteases. Of these, 62.3% induced biofilm with proteinase-3, 15% with cathepsin G, 10% with cathepsin B and 5% with MMP -9, where most of the protease-induced biofilm isolates were commensal strains (skin). In the biofilm formation kinetics analysis, the addition of phenylmethylsulfonyl fluoride (PMSF; a proteinase-3 inhibitor) showed that proteinase-3 participates in the cell aggregation stage of biofilm formation. A biofilm induced with proteinase-3 and DNAse-treated significantly reduced biofilm formation at an early time (initial adhesion stage of biofilm formation) compared to untreated proteinase-3-induced biofilm (p < 0.05). A catheter inoculated with a commensal (skin) non-biofilm-forming S. epidermidis isolate treated with proteinase-3 and another one without the enzyme were inserted into the back of a mouse. After 7 days of incubation period, the catheters were recovered and the number of grown bacteria was quantified, finding a higher amount of adhered proteinase-3-treated bacteria in the catheter than non-proteinase-3-treated bacteria (p < 0.05). Commensal non-biofilm-forming S. epidermidis in the presence of neutrophil cells significantly induced the biofilm formation when multiplicity of infection (MOI) 1:0.01 (neutrophil:bacteria) was used, but the addition of a cocktail of protease inhibitors impeded biofilm formation. A neutrophil:bacteria assay did not induce neutrophil extracellular traps (NETs). Our results suggest that neutrophils, in the presence of commensal non-biofilm-forming S. epidermidis, do not generate NETs formation. The effect of neutrophils is the production of proteases, and proteinase-3 releases bacterial DNA at the initial adhesion, favoring cell aggregation and subsequently leading to biofilm formation.
Collapse
|
59
|
Gaio V, Lima T, Vilanova M, Cerca N, França A. mazEF Homologue Has a Minor Role in Staphylococcus epidermidis 1457 Virulence Potential. Front Cell Infect Microbiol 2022; 11:803134. [PMID: 35096651 PMCID: PMC8792614 DOI: 10.3389/fcimb.2021.803134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus epidermidis biofilm cells are characterized by increased antimicrobial tolerance and improved ability to evade host immune system defenses. These features are, in part, due to the presence of viable but non-culturable (VBNC) cells. A previous study identified genes potentially involved in VBNC cells formation in S. epidermidis biofilms, among which SERP1682/1681 raised special interest due to their putative role as a toxin–antitoxin system of the mazEF family. Herein, we constructed an S. epidermidis mutant lacking the mazEF genes homologues and determined their role in (i) VBNC state induction during biofilm formation, (ii) antimicrobial susceptibility, (iii) survival in human blood and plasma, and (iv) activation of immune cells. Our results revealed that mazEF homologue did not affect the proportion of VBNC cells in S. epidermidis 1457, refuting the previous hypothesis that mazEF homologue could be linked with the emergence of VBNC cells in S. epidermidis biofilms. Additionally, mazEF homologue did not seem to influence key virulence factors on this strain, since its deletion did not significantly affect the mutant biofilm formation capacity, antimicrobial tolerance or the response by immune cells. Surprisingly, our data suggest that mazEF does not behave as a toxin–antitoxin system in S. epidermidis strain 1457, since no decrease in the viability and culturability of bacteria was found when only the mazF toxin homologue was being expressed.
Collapse
Affiliation(s)
- Vânia Gaio
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Tânia Lima
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Manuel Vilanova
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Nuno Cerca
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Angela França
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, Braga, Portugal
| |
Collapse
|
60
|
Elashnikov R, Rimpelová S, Lyutakov O, Pavlíčková VS, Khrystonko O, Kolská Z, Švorčík V. Ciprofloxacin-Loaded Poly( N-isopropylacrylamide- co-acrylamide)/Polycaprolactone Nanofibers as Dual Thermo- and pH-Responsive Antibacterial Materials. ACS APPLIED BIO MATERIALS 2022; 5:1700-1709. [DOI: 10.1021/acsabm.2c00069] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Roman Elashnikov
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Oleksiy Lyutakov
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Vladimíra Svobodová Pavlíčková
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Olena Khrystonko
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Zdeňka Kolská
- Materials Centre, Faculty of Science, J. E. Purkyně University, Pasteurova 15, 40096 Ústí nad Labem, Czech Republic
| | - Václav Švorčík
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
61
|
Sethi G, Sethi S, Krishna R. Multi-epitope based vaccine design against Staphylococcus epidermidis: A subtractive proteomics and immunoinformatics approach. Microb Pathog 2022; 165:105484. [PMID: 35301068 DOI: 10.1016/j.micpath.2022.105484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Staphylococcus epidermidis has emerged as a major contributor of nosocomial infections across the world. With the increased rate of emerging resistant and previously undefined infectious diseases, there is a growing need to develop a novel vaccine possessing required immunogenic properties. The adopted reverse vaccinology approach identified "IMPNQILTI" of LysM domain protein, "YSYTYTIDA" of staphylococcal secretory antigen SsaA, and "YNYDANTGQ" neutral metalloproteinaseas potential peptides for vaccine design. The 9-mer epitope of target proteins is antigenic, virulent, surface-exposed, non-allergenic, and conserved across various strains of S. epidermidis. Protein-protein interactions study indicated the involvement of target proteins in major biological pathways for S. epidermidis pathogenesis. Protein-peptide docking was performed, and population coverage analysis showed significant interactions of T-cell epitopes with the HLA-binding molecules while covering 90.58% of the world's population. Further, a multi-epitope vaccine of 177 amino acids long was constructed. Docking with Toll-like receptor (TLR-2) molecule confirmed the effective interaction of the vaccine with the receptor. The vaccine efficiency in generating an effective immune response in the host was evaluated by immune simulation. Finally, in silico cloning confirmed that the constructed vaccine can be efficiently expressed in E. coli. However, the designed vaccine needs experimental validation to determine the effectiveness and immunogenicity profile, which will ensure an active immunity against S. epidermidis.
Collapse
Affiliation(s)
- Guneswar Sethi
- Department of Bioinformatics, Pondicherry University, Puducherry, 605014, India.
| | - Satyanarayan Sethi
- Central Institute of Freshwater Aquaculture, Research Institute, Bhubaneswar, 751002, India.
| | - Ramadas Krishna
- Department of Bioinformatics, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
62
|
Mirzaei R, Alikhani MY, Arciola CR, Sedighi I, Yousefimashouf R, Bagheri KP. Prevention, inhibition, and degradation effects of melittin alone and in combination with vancomycin and rifampin against strong biofilm producer strains of methicillin-resistant Staphylococcus epidermidis. Biomed Pharmacother 2022; 147:112670. [PMID: 35123230 DOI: 10.1016/j.biopha.2022.112670] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus epidermidis (MRSE) bacteria are being recognized as true pathogens as they are able to resist methicillin and commonly form biofilms. Recent studies have shown that antimicrobial peptides (AMPs) are promising agents against biofilm-associated bacterial infections. In this study, we aimed to explore the antibiofilm activity of melittin, either alone or in combination with vancomycin and rifampin, against biofilm-producing MRSE strains. Minimum biofilm preventive concentration (MBPC), minimum biofilm inhibition concentration (MBIC), and minimum biofilm eradication concentration (MBEC), as well as fractional biofilm preventive-, inhibitory-, and eradication concentrations (FBPCi, FBICi, and FBECi), were determined for the antimicrobial agents tested. Cytotoxicity and hemolytic activity of melittin at its synergistic concentration were examined on human embryonic kidney cells (HEK-293) and Red Blood Cells (RBCs), respectively. The effect of melittin on the downregulation of biofilm-associated genes was explored using Real-Time PCR. MBPC, MBIC, and MBEC values for melittin were in the range of 0.625-20, 0.625-20, and 10-40 μg/μL, respectively. Melittin showed high synergy (FBPCi, FBICi and FBECi < 0.5). The synergism resulted in a 64-512-fold, 2-16 and 2-8-fold reduction in melittin, rifampicin and vancomycin concentrations, respectively. The synergistic melittin concentration found to be effective did not manifest either cytotoxicity on HEK-293 or hemolytic activity on RBCs. Results showed that melittin downregulated the expression of biofilm-associated icaA, aap, and psm genes in all isolates tested, ranging from 0.04-folds to 2.11-folds for icaA and from 0.05 to 3.76-folds for aap and psm. The preventive and therapeutic indexes of melittin were improved 8-fold when combined with vancomycin and rifampin. Based on these findings, the combination of melittin with conventional antibiotics could be proposed for treating or preventing biofilm-associated MRSE infections.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Carla Renata Arciola
- Laboratorio di Patologia delle Infezioni Associate all'Impianto IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Iraj Sedighi
- Department of Pediatrics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
63
|
Witty M. Examples of potato epidermis endophytes and rhizosphere microbes that may be human pathogens contributing to potato peel colic. ACTA ALIMENTARIA 2022. [DOI: 10.1556/066.2021.00157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
Potato tubers defend themselves against herbivores with endogenous secondary compounds such as solanine and scopolamine. They also recruit endophytes and members of the tuberosphere to repel herbivores. Many of these endophyte defence features are overcome by cooking, with some notable exceptions that have been identified by rDNA analysis of potato peel samples and may account for some previously unrecognised features of potato peel colic. This is relevant regarding the rather modern way of cooking, where the potato peel is left intact in food and consumed.
Collapse
Affiliation(s)
- M. Witty
- Math and Science Department, School of Pure and Applied Sciences, Florida SouthWestern State College, 8099 College Parkway, Fort Myers, Florida 33919, USA
| |
Collapse
|
64
|
Antimicrobial characterization of a titanium coating derived from mussel-glue and Bothrops asper snake venom for the prevention of implant-associated infections caused by Staphylococcus. ELECTRON J BIOTECHN 2022. [DOI: 10.1016/j.ejbt.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
65
|
Nadar S, Khan T, Patching SG, Omri A. Development of Antibiofilm Therapeutics Strategies to Overcome Antimicrobial Drug Resistance. Microorganisms 2022; 10:microorganisms10020303. [PMID: 35208758 PMCID: PMC8879831 DOI: 10.3390/microorganisms10020303] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
A biofilm is a community of stable microorganisms encapsulated in an extracellular matrix produced by themselves. Many types of microorganisms that are found on living hosts or in the environment can form biofilms. These include pathogenic bacteria that can serve as a reservoir for persistent infections, and are culpable for leading to a broad spectrum of chronic illnesses and emergence of antibiotic resistance making them difficult to be treated. The absence of biofilm-targeting antibiotics in the drug discovery pipeline indicates an unmet opportunity for designing new biofilm inhibitors as antimicrobial agents using various strategies and targeting distinct stages of biofilm formation. The strategies available to control biofilm formation include targeting the enzymes and proteins specific to the microorganism and those involved in the adhesion pathways leading to formation of resistant biofilms. This review primarily focuses on the recent strategies and advances responsible for identifying a myriad of antibiofilm agents and their mechanism of biofilm inhibition, including extracellular polymeric substance synthesis inhibitors, adhesion inhibitors, quorum sensing inhibitors, efflux pump inhibitors, and cyclic diguanylate inhibitors. Furthermore, we present the structure–activity relationships (SAR) of these agents, including recently discovered biofilm inhibitors, nature-derived bioactive scaffolds, synthetic small molecules, antimicrobial peptides, bioactive compounds isolated from fungi, non-proteinogenic amino acids and antibiotics. We hope to fuel interest and focus research efforts on the development of agents targeting the uniquely complex, physical and chemical heterogeneous biofilms through a multipronged approach and combinatorial therapeutics for a more effective control and management of biofilms across diseases.
Collapse
Affiliation(s)
- Sahaya Nadar
- Department of Pharmaceutical Chemistry, St. John Institute of Pharmacy and Research, Mumbai 400056, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India;
| | - Simon G. Patching
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: or (S.G.P.); (A.O.)
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: or (S.G.P.); (A.O.)
| |
Collapse
|
66
|
Thill P, Robineau O, Roosen G, Patoz P, Gachet B, Lafon-Desmurs B, Tetart M, Nadji S, Senneville E, Blondiaux N. Rifabutin versus rifampicin bactericidal and antibiofilm activities against clinical strains of Staphylococcus spp. isolated from bone and joint infections. J Antimicrob Chemother 2022; 77:1036-1040. [PMID: 35028671 DOI: 10.1093/jac/dkab486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/08/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Staphylococci account for approximately 60% of periprosthetic joint infections (PJIs). Rifampicin (RMP) combination therapy is generally considered to be the treatment of choice for staphylococcal PJIs but carries an important risk of adverse events and drug-drug interactions. Rifabutin (RFB) shares many of the properties of rifampicin but causes fewer adverse events. OBJECTIVES To compare the minimal inhibitory concentration (MIC), the minimum bactericidal concentrations (MBC), and the minimum biofilm eradication concentrations (MBEC) of rifabutin and rifampicin for staphylococcal clinical strains isolated from PJIs. METHODS 132 clinical strains of rifampicin-susceptible staphylococci [51 Staphylococcus aureus (SA), 48 Staphylococcus epidermidis (SE) and 33 other coagulase-negative staphylococci (CoNS)] were studied. The MBC and the MBEC were determined using the MBEC® Assay for rifabutin and rifampicin and were compared. RESULTS When compared with the rifampicin MIC median value, the rifabutin MIC median value was significantly higher for SA (P < 0.05), but there was no statistically significant difference for SE (P = 0.25) and CoNS (P = 0.29). The rifabutin MBC median value was significantly higher than that of rifampicin for SA (P = 0.003) and was lower for SE (P = 0.003) and CoNS (P = 0.03). Rifabutin MBEC median value was statistically lower than that of rifampicin for all strains tested. CONCLUSIONS Using the determination of MBEC values, our study suggests that rifabutin is more effective than rifampicin against clinical strains of Staphylococcus spp. obtained from PJIs. Using MBECs instead of MICs seems to be of interest when considering biofilms. In vivo higher efficacy of rifabutin when compared with rifampicin needs to be confirmed.
Collapse
Affiliation(s)
- Pauline Thill
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France
| | - Olivier Robineau
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | - Gabrielle Roosen
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Pierre Patoz
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Benoit Gachet
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | | | - Macha Tetart
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France
| | - Safia Nadji
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Eric Senneville
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | - Nicolas Blondiaux
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France.,Univ. Lille, CNRS, Inserm, Institut Pasteur de Lille, U1019 - UMR9017 Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
67
|
Zwicker P, Schmidt T, Hornschuh M, Lode H, Kramer A, Müller G. In vitro response of THP-1 derived macrophages to antimicrobially effective PHMB-coated Ti6Al4V alloy implant material with and without contamination with S. epidermidis and P. aeruginosa. Biomater Res 2022; 26:1. [PMID: 35000621 PMCID: PMC8744236 DOI: 10.1186/s40824-021-00247-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/05/2021] [Indexed: 12/23/2022] Open
Abstract
AIM Periprosthetic joint infections are a devastating complication after arthroplasty, leading to rejection of the prosthesis. The prevention of septic loosening may be possible by an antimicrobial coating of the implant surface. Poly (hexamethylene) biguanide hydrochloride [PHMB] seems to be a suitable antiseptic agent for this purpose since previous studies revealed a low cytotoxicity and a long-lasting microbicidal effect of Ti6Al4V alloy coated with PHMB. To preclude an excessive activation of the immune system, possible inflammatory effects on macrophages upon contact with PHMB-coated surfaces alone and after killing of S. epidermidis and P. aeruginosa are analyzed. METHODS THP-1 monocytes were differentiated to M0 macrophages by phorbol 12-myristate 13-acetate and seeded onto Ti6Al4V surfaces coated with various amounts of PHMB. Next to microscopic immunofluorescence analysis of labeled macrophages after adhesion on the coated surface, measurement of intracellular reactive oxygen species and analysis of cytokine secretion at different time points without and with previous bacterial contamination were conducted. RESULTS No influence on morphology of macrophages and only slight increases in iROS generation were detected. The cytokine secretion pattern depends on the surface treatment procedure and the amount of adsorbed PHMB. The PHMB coating resulted in a high reduction of viable bacteria, resulting in no significant differences in cytokine secretion as reaction to coated surfaces with and without bacterial burden. CONCLUSION Ti6Al4V specimens after alkaline treatment followed by coating with 5-7 μg PHMB and specimens treated with H2O2 before PHMB-coating (4 μg) had the smallest influence on the macrophage phienotype and thus are considered as the surface with the best cytocompatibility to macrophages tested in the present study.
Collapse
Affiliation(s)
- Paula Zwicker
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany.
| | - Thomas Schmidt
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Melanie Hornschuh
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Holger Lode
- Department of Pediatric Hematology and Oncology, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Axel Kramer
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Gerald Müller
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| |
Collapse
|
68
|
da Silva Filho PM, Andrade AL, Lopes JBAC, Pinheiro ADA, de Vasconcelos MA, Fonseca SGDC, Lopes LGDF, Sousa EHS, Teixeira EH, Longhinotti E. The biofilm inhibition activity of a NO donor nanosilica with enhanced antibiotics action. Int J Pharm 2021; 610:121220. [PMID: 34687814 DOI: 10.1016/j.ijpharm.2021.121220] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/30/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) has emerged as a promising antibacterial agent, where NO donor compounds have been explored. Here, we investigated the role of a silica nanoparticle containing nitroprusside (MPSi-NP) as a NO donor agent against methicillin-sensitive (ATCC 25,923 and ATCC 12228) and methicillin-resistant (ATCC 700,698 and ATCC 35984) Staphylococcus strains. Biofilm inhibition was studied along with antibiotic activity in combination with standard antibiotics (ampicillin and tetracycline). MPSi-NP exhibited thermal release of 63% of NO within 24 h, while free nitroprusside released only 18% during a dialysis assay, indicating an assisted release of NO mediated by the nanoparticles. This nanomaterial showed only a moderate activity in blocking biofilm production, but exhibited a significant decrease in the number of viable bacterial cells (over 600-fold for Staphylococcus aureus ATCC 700,698 and Staphylococcus epidermidis ATCC 35984). Remarkably, even using MPSi-NP at concentrations below any antibacterial action, its combination with ampicillin promoted a significant decrease in MIC for resistant strains of S. aureus ATCC 700,698 (2-fold) and S. epidermidis ATCC 35,984 (4-fold). A carbopol-based gel formulation with MPSi-NP (0.5% w/w) was prepared and showed a zone of inhibition of 7.7 ± 0.6 mm for S. epidermidis ATCC 35984. Topical use of MPSi-NP in combination with antibiotics might be a manageable strategy to prevent and eventually treat complicated resistant bacterial infections.
Collapse
Affiliation(s)
- Pedro Martins da Silva Filho
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, Fortaleza, CE 60440-900, Brazil; Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, Fortaleza, CE 60440-900, Brazil
| | - Alexandre Lopes Andrade
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil
| | - Jessica Barros Arrais Cruz Lopes
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil
| | - Aryane de Azevedo Pinheiro
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil
| | - Mayron Alves de Vasconcelos
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil; Departamento de Ciências Biológicas, Faculdade de Ciências Exatas e Naturais, Universidade do Estado do Rio Grande do Norte, Mossoró, RN 59610-090, Brazil; Universidade do Estado de Minas Gerais, Unidade de Divinópolis, Divinópolis, MG 35501-170, Brazil
| | | | - Luiz Gonzaga de França Lopes
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, Fortaleza, CE 60440-900, Brazil
| | - Eduardo Henrique Silva Sousa
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, Fortaleza, CE 60440-900, Brazil.
| | - Edson Holanda Teixeira
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil.
| | - Elisane Longhinotti
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, Fortaleza, CE 60440-900, Brazil.
| |
Collapse
|
69
|
Suneel Kumar A, Smiline Girija AS, Naga Srilatha B. Characterization of biofilm producing methicillin resistant coagulase negative Staphylococci from India. Acta Microbiol Immunol Hung 2021; 69:35-40. [PMID: 34898472 DOI: 10.1556/030.2021.01538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/26/2021] [Indexed: 01/12/2023]
Abstract
Methicillin-resistant coagulase-negative staphylococci (MR-CoNS) cause infectious diseases due to their potential to form biofilm and further colonization in hospital materials. This study evaluated the antibiotic susceptible phenotypes, biofilm-producing ability, and biofilm-associated genes (mecA, icaAD, bap, cna, and fnbA). Biofilm formation was detected through Congo red agar (CRA) method and MTP method. The presence of biofilm and associated genes in MR-CoNS were detected by PCR. A total of 310 (55.95%) isolates produced the biofilm. Among these isolates, Staphylococcus haemolyticus (34.83%), Staphylococcus epidermis (31.93%), Staphylococcus capitis (16.77%), Staphylococcus cohnii (10.96%), and Staphylococcus hominis (5.48%) were identified. The antimicrobial susceptibility pattern of CoNS isolates indicated resistance to cefoxitin (100%), erythromycin (94.8%), ciprofloxacin (66.7%), sulfamethoxazole/trimethoprim (66.7%), gentamicin (66.12%), and clindamycin (62.9%). Resistance rate to mupirocin was 48.5% in S. epidermidis and 38.9% in S. haemolyticus isolates. All isolates were sensitive to vancomycin and linezolid. The prevalence rates of icaAD, bap, fnbA, and cna were 18.06%, 12.5%, 47.4%, and 27.4%, respectively. icaAD and bap genes were detected in 18.06% and 12.5% of MR-CoNS isolates. fnbA and cna genes were detected in 47.41% and 27.41% of MRCoNS isolates. icaAD positive strains exhibited a significant increase in the biofilm formation compared with those that lacked icaAD (0.86 (0.42, 1.39) versus 0.36 (0.14, 0.75), respectively; P < 0.001). In conclusion, the majority of MR-CoNS isolates were biofilm producers, and S. capitis, which possessed icaAD genes, ranked as the great biofilm producer than other Staphylococcus. The study's findings are important to form a strategy to control biofilm formation as an alternative strategy to counter the spread of MR-CoNS in healthcare settings.
Collapse
Affiliation(s)
- A Suneel Kumar
- 1 Department of Microbiology, Rajiv Gandhi Institute of Medical Sciences (RIMS), Government Medical College & Hospital, Kadapa, Andhra Pradesh, India
- 2 Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - A S Smiline Girija
- 2 Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - B Naga Srilatha
- 1 Department of Microbiology, Rajiv Gandhi Institute of Medical Sciences (RIMS), Government Medical College & Hospital, Kadapa, Andhra Pradesh, India
| |
Collapse
|
70
|
Pandit S, Li M, Chen Y, Rahimi S, Mokkapati V, Merlo A, Yurgens A, Mijakovic I. Graphene-Based Sensor for Detection of Bacterial Pathogens. SENSORS 2021; 21:s21238085. [PMID: 34884089 PMCID: PMC8662450 DOI: 10.3390/s21238085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 11/23/2022]
Abstract
Microbial colonization to biomedical surfaces and biofilm formation is one of the key challenges in the medical field. Recalcitrant biofilms on such surfaces cause serious infections which are difficult to treat using antimicrobial agents, due to their complex structure. Early detection of microbial colonization and monitoring of biofilm growth could turn the tide by providing timely guidance for treatment or replacement of biomedical devices. Hence, there is a need for sensors, which could generate rapid signals upon bacterial colonization. In this study, we developed a simple prototype sensor based on pristine, non-functionalized graphene. The detection principle is a change in electrical resistance of graphene upon exposure to bacterial cells. Without functionalization with specific receptors, such sensors cannot be expected to be selective to certain bacteria. However, we demonstrated that two different bacterial species can be detected and differentiated by our sensor due to their different growth dynamics, adherence pattern, density of adhered bacteria and microcolonies formation. These distinct behaviors of tested bacteria depicted distinguishable pattern of resistance change, resistance versus gate voltage plot and hysteresis effect. This sensor is simple to fabricate, can easily be miniaturized, and can be effective in cases when precise identification of species is not needed.
Collapse
Affiliation(s)
- Santosh Pandit
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; (S.P.); (Y.C.); (S.R.); (V.M.); (A.M.)
| | - Mengyue Li
- Department of Microtechnology and Nanoscience, Chalmers University of Technology, 412 96 Göteborg, Sweden; (M.L.); (A.Y.)
| | - Yanyan Chen
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; (S.P.); (Y.C.); (S.R.); (V.M.); (A.M.)
| | - Shadi Rahimi
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; (S.P.); (Y.C.); (S.R.); (V.M.); (A.M.)
| | - Vrss Mokkapati
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; (S.P.); (Y.C.); (S.R.); (V.M.); (A.M.)
| | - Alessandra Merlo
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; (S.P.); (Y.C.); (S.R.); (V.M.); (A.M.)
| | - August Yurgens
- Department of Microtechnology and Nanoscience, Chalmers University of Technology, 412 96 Göteborg, Sweden; (M.L.); (A.Y.)
| | - Ivan Mijakovic
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden; (S.P.); (Y.C.); (S.R.); (V.M.); (A.M.)
- Novo Nordisk Foundation, Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Correspondence: ; Tel.: +46-(0)7-0982-8446
| |
Collapse
|
71
|
Oliveira F, Rohde H, Vilanova M, Cerca N. Fighting Staphylococcus epidermidis Biofilm-Associated Infections: Can Iron Be the Key to Success? Front Cell Infect Microbiol 2021; 11:798563. [PMID: 34917520 PMCID: PMC8670311 DOI: 10.3389/fcimb.2021.798563] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus epidermidis is one of the most important commensal microorganisms of human skin and mucosae. However, this bacterial species is also the cause of severe infections in immunocompromised patients, specially associated with the utilization of indwelling medical devices, that often serve as a scaffold for biofilm formation. S. epidermidis strains are often multidrug resistant and its association with biofilm formation makes these infections hard to treat. Their remarkable ability to form biofilms is widely regarded as its major pathogenic determinant. Although a significant amount of knowledge on its biofilm formation mechanisms has been achieved, we still do not understand how the species survives when exposed to the host harsh environment during invasion. A previous RNA-seq study highlighted that iron-metabolism associated genes were the most up-regulated bacterial genes upon contact with human blood, which suggested that iron acquisition plays an important role in S. epidermidis biofilm development and escape from the host innate immune system. In this perspective article, we review the available literature on the role of iron metabolism on S. epidermidis pathogenesis and propose that exploiting its dependence on iron could be pursued as a viable therapeutic alternative.
Collapse
Affiliation(s)
- Fernando Oliveira
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Holger Rohde
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel Vilanova
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Nuno Cerca
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| |
Collapse
|
72
|
The Vancomycin Resistance-Associated Regulatory System VraSR Modulates Biofilm Formation of Staphylococcus epidermidis in an ica-Dependent Manner. mSphere 2021; 6:e0064121. [PMID: 34550006 PMCID: PMC8550092 DOI: 10.1128/msphere.00641-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The two-component system VraSR responds to the cell wall-active antibiotic stress in Staphylococcus epidermidis. To study its regulatory function in biofilm formation, a vraSR deletion mutant (ΔvraSR) was constructed using S. epidermidis strain 1457 (SE1457) as the parent strain. Compared to SE1457, the ΔvraSR mutant showed impaired biofilm formation both in vitro and in vivo with a higher ratio of dead cells within the biofilm. Consistently, the ΔvraSR mutant produced much less polysaccharide intercellular adhesin (PIA). The ΔvraSR mutant also showed increased susceptibility to the cell wall inhibitor and SDS, and its cell wall observed under a transmission electron microscope (TEM) appeared to be thinner and interrupted, which is in accordance with higher susceptibility to the stress. Complementation of vraSR in the ΔvraSR mutant restored the biofilm formation and the cell wall thickness to wild-type levels. Transcriptome sequencing (RNA-Seq) showed that the vraSR deletion affected the transcription levels of 73 genes, including genes involved in biofilm formation, bacterial programmed cell death (CidA-LrgAB system), glycolysis/gluconeogenesis, the pentose phosphate pathway (PPP), and the tricarboxylic acid (TCA) cycle, etc. The results of RNA-Seq were confirmed by quantitative real-time reverse transcription-PCR (qRT-PCR). In the ΔvraSR mutant, the expression of icaA and lrgAB was downregulated and the expression of icaR and cidA was upregulated, in comparison to that of SE1457. The transcriptional levels of antibiotic-resistant genes (pbp2, serp1412, murAA, etc.) had no significant changes. An electrophoretic mobility shift assay further revealed that phosphorylated VraR bound to the promoter regions of the ica operon, as well as its own promoter region. This study demonstrates that in S. epidermidis, VraSR is an autoregulator and directly regulates biofilm formation in an ica-dependent manner. Upon cell wall stress, it indirectly regulates cell death and drug resistance in association with alterations to multiple metabolism pathways. IMPORTANCES. epidermidis is a leading cause of hospital-acquired catheter-related infections, and its pathogenicity depends mostly on its ability to form biofilms on implants. The biofilm formation is a complex procedure that involves multiple regulating factors. Here, we show that a vancomycin resistance-associated two-component regulatory system, VraSR, plays an important role in modulating S. epidermidis biofilm formation and tolerance to stress. We demonstrate that S. epidermidis VraSR is an autoregulated system that selectively responds to stress targeting cell wall synthesis. Besides, phosphorylated VraR can bind to the promoter region of the ica operon and directly regulates polysaccharide intercellular adhesin production and biofilm formation in S. epidermidis. Furthermore, VraSR may indirectly modulate bacterial cell death and extracellular DNA (eDNA) release in biofilms through the CidA-LrgAB system. This work provides a new molecular insight into the mechanisms of VraSR-mediated modulation of the biofilm formation and cell death of S. epidermidis.
Collapse
|
73
|
Introducing image-guided synovial aspiration and biopsy in assessing peri-prosthetic joint infection: an early single-centre experience. Skeletal Radiol 2021; 50:2031-2040. [PMID: 33825021 DOI: 10.1007/s00256-021-03774-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 02/02/2023]
Abstract
AIM Synovial sampling can be used in the diagnosis of peri-prosthetic joint infection (PJI). The purpose of this study was to establish the role of simultaneous image-guided synovial aspiration and biopsy (SAB) during an initial 2-year experience at our institution. METHODS Retrospective review of consecutive SABs performed during 2014-2016 at a tertiary referral musculoskeletal centre. Radiological SAB microbiology culture results were compared with intra-operative surgical samples or multidisciplinary team (MDT) meeting outcome at 1-year follow-up if surgery was not undertaken. Sensitivity, specificity and accuracy of synovial aspiration (SA), synovial biopsy (SB) and simultaneous SAB were calculated. RESULTS 103 patients (46 male, 57 female) totalling 111 procedures were analysed with mean age 65 years (range 31-83). Image-guided synovial procedures were performed on 52 (46.9%) hip and 59 (53.1%) knee joint prostheses. The mean combined sensitivity, specificity and accuracy for the entire cohort was 72.6%, 96.9% and 90%, respectively. When only SB was obtained, diagnostic accuracy (92.5%) was similar to SA alone (94.1%). In total, there were 21 (18.9%) true-positive, 80 (72.1%) true-negative, 2 (1.8%) false-positive and 8 (7.2%) false-negative cases (PPV 91.3% and NPV 90.9%). No post-procedural complications were recorded at 1-year follow-up. CONCLUSION Percutaneous image-guided SAB is a valuable technique in assessing suspected PJI, with most samples indicative of infective status and causative organisms when validated against intra-operative results and specialist MDT evaluation. Image-guided SB is a safe and useful additional procedure following failed SA with equivalent levels of diagnostic accuracy.
Collapse
|
74
|
Elhakim YA, Ali AE, Hosny AEDMS, Abdeltawab NF. Zinc Deprivation as a Promising Approach for Combating Methicillin-Resistant Staphylococcus aureus: A Pilot Study. Pathogens 2021; 10:1228. [PMID: 34684179 PMCID: PMC8540720 DOI: 10.3390/pathogens10101228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/06/2021] [Accepted: 09/15/2021] [Indexed: 12/23/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are a global health burden with an urgent need for antimicrobial agents. Studies have shown that host immune responses limit essential metals such as zinc during infection, leading to the limitation of bacterial virulence. Thus, the deprivation of zinc as an important co-factor for the activity of many S. aureus enzymes can be a potential antimicrobial approach. However, the effect of zinc deprivation on S. aureus and MRSA is not fully understood. Therefore, the current study aimed to dissect the effects of zinc deprivation on S. aureus hemolytic activity and biofilm formation through employing biochemical and genetic approaches to study the effect of zinc deprivation on S. aureus growth and virulence. Chemically defined media (CDM) with and without ZnCl2, was used to assess the effect of zinc deprivation on growth, biofilm formation, and hemolytic activity in methicillin-susceptible S. aureus (MSSA) RN6390 and MRSA N315 strains. Zinc deprivation decreased the growth of RN6390 and N315 S. aureus strains significantly by 1.5-2 folds, respectively compared to the zinc physiological range encountered by the bacteria in the human body (7-20 µM) (p < 0.05). Zinc deprivation significantly reduced biofilm formation by 1.5 folds compared to physiological levels (p < 0.05). Moreover, the hemolytic activity of RN6390 and N315 S. aureus strains was significantly decreased by 20 and 30 percent, respectively compared to physiological zinc levels (p < 0.05). Expression of biofilm-associated transcripts levels at late stage of biofilm formation (20 h) murein hydrolase activator A (cidA) and cidB were downregulated by 3 and 5 folds, respectively (p < 0.05) suggested an effect on extracellular DNA production. Expression of hemolysins-associated genes (hld, hlb, hla) was downregulated by 3, 5, and 10 folds, respectively, in absence of zinc (p < 0.001). Collectively the current study showed that zinc deprivation in vitro affected growth, biofilm formation, and hemolytic activity of S. aureus. Our in vitro findings suggested that zinc deprivation can be a potential supportive anti-biofilm formation and antihemolytic approach to contain MRSA topical infections.
Collapse
Affiliation(s)
- Yomna A. Elhakim
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (Y.A.E.); (A.E.-D.M.S.H.)
| | - Amal E. Ali
- Department of Microbiology and Immunology, Faculty of Pharmacy, Future University in Egypt, New Cairo 12311, Egypt;
| | - Alaa El-Dien M. S. Hosny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (Y.A.E.); (A.E.-D.M.S.H.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 12055, Egypt
| | - Nourtan F. Abdeltawab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (Y.A.E.); (A.E.-D.M.S.H.)
| |
Collapse
|
75
|
Chantraine C, Mathelié-Guinlet M, Pietrocola G, Speziale P, Dufrêne YF. AFM Identifies a Protein Complex Involved in Pathogen Adhesion Which Ruptures at Three Nanonewtons. NANO LETTERS 2021; 21:7595-7601. [PMID: 34469164 DOI: 10.1021/acs.nanolett.1c02105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Staphylococci bind to the blood protein von Willebrand Factor (vWF), thereby causing endovascular infections. Whether and how this interaction occurs with the medically important pathogen Staphylococcus epidermidis is unknown. Using single-molecule experiments, we demonstrate that the S. epidermidis protein Aap binds vWF via an ultrastrong force, ∼3 nN, the strongest noncovalent biological bond ever reported, and we show that this interaction is activated by tensile loading, suggesting a catch-bond behavior. Aap-vWF binding involves exclusively the A1 domain of vWF but requires both the A and B domains of Aap, as revealed by inhibition assays using specific monoclonal antibodies. Collectively, our results point to a mechanism where force-induced unfolding of the B repeats activates the A domain of Aap, shifting it from a weak- to a strong-binding state, which then engages into an ultrastrong interaction with vWF A1. This shear-dependent function of Aap offers promise for innovative antistaphylococcal therapies.
Collapse
Affiliation(s)
- Constance Chantraine
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Marion Mathelié-Guinlet
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Giampiero Pietrocola
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Pietro Speziale
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
76
|
Svensson Malchau K, Tillander J, Zaborowska M, Hoffman M, Lasa I, Thomsen P, Malchau H, Rolfson O, Trobos M. Biofilm properties in relation to treatment outcome in patients with first-time periprosthetic hip or knee joint infection. J Orthop Translat 2021; 30:31-40. [PMID: 34485075 PMCID: PMC8385121 DOI: 10.1016/j.jot.2021.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/27/2021] [Accepted: 05/31/2021] [Indexed: 11/15/2022] Open
Abstract
Background Periprosthetic joint infections (PJI) are challenging complications following arthroplasty. Staphylococci are a frequent cause of PJI and known biofilm producers. Biofilm formation decreases antimicrobial susceptibility, thereby challenging favourable treatment outcomes. The aims of this study were to characterize the biofilm abilities and antimicrobial susceptibilities of staphylococci causing first-time PJI and correlate them to clinical outcome (infection resolution and recurrence). Methods Reoperations for PJI of the hip or knee between 1st January 2012 to 30th June 2015 performed at the Sahlgrenska University Hospital were identified in a local database. Medical records were reviewed and clinical parameters recorded for patients whose intraoperative bacterial isolates had been stored at the clinical laboratory. Staphylococcal strains isolated from reoperations due to first-time PJI were characterised by their ability to form biofilms using the microtiter plate test. Antimicrobial susceptibility of the strains was determined by minimum inhibitory concentration (MIC) when grown planktonically, and by minimum biofilm eradication concentration (MBEC) when grown as biofilms. MBEC determination was conducted using the Calgary biofilm device (CBD) and a custom-made antimicrobial susceptibility plate containing eight clinically relevant antimicrobial agents. Results The study group included 49 patients (70 bacterial strains) from first-time PJI, whereof 24 (49%) patients had recurrent infection. Strong biofilm production was significantly associated with recurrent infection. Patients infected with strong biofilm producers had a five-fold increased risk for recurrent infection. Strains grown as biofilms were over 8000 times more resistant to antimicrobial agents compared to planktonic cultures. Biofilms were more susceptible to rifampicin compared to other antimicrobials in the assay. Increased biofilm susceptibility (MBEC > MIC) was observed for the majority of the bacterial strains and antimicrobial agents. Conclusions Strong biofilm production was significantly associated with increased antimicrobial resistance and PJI recurrence. This underscores the importance of determining biofilm production and susceptibility as part of routine diagnostics in PJI. Strong staphylococcal biofilm production may have implications on therapeutic choices and suggest more extensive surgery. Furthermore, despite the increased biofilm resistance to rifampicin, results from this study support its use in staphylococcal PJI. The Translational Potential of this Article Like for many biomaterial-associated infections, staphylococci are a common cause of PJI. Their ability to adhere to surfaces and produce biofilms on medical devices is proposed to play a role. However, clinical studies where biofilm properties are directly linked to patient outcome are scarce. This study demonstrates that the majority of staphylococci isolated from first-time PJI were biofilm producers with increased antimicrobial resistance. Patients suffering an infection caused by a staphylococcal strain with strong biofilm production ability had a five-fold greater risk of recurrent infection. This novel finding suggests the importance of evaluating biofilm production as a diagnostic procedure for the guidance of treatment decisions in PJI.
Collapse
Affiliation(s)
- Karin Svensson Malchau
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Jonatan Tillander
- Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magdalena Zaborowska
- Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden.,Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria Hoffman
- Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden.,Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Iñigo Lasa
- Microbial Pathogenesis Research Unit, Public University of Navarre, Pamplona, Spain
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Malchau
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Ola Rolfson
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Margarita Trobos
- Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden.,Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
77
|
Kia C, Cusano A, Messina J, Muench LN, Chadayammuri V, McCarthy MB, Umejiego E, Mazzocca AD. Effectiveness of topical adjuvants in reducing biofilm formation on orthopedic implants: an in vitro analysis. J Shoulder Elbow Surg 2021; 30:2177-2183. [PMID: 33529773 DOI: 10.1016/j.jse.2020.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/27/2020] [Accepted: 12/05/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND HYPOTHESIS The treatment of periprosthetic joint infection is complicated by the presence of residual biofilm, which resists eradication owing to bacterial adherence to orthopedic implants. The purpose of this study was to compare Bactisure (Zimmer Biomet, Warsaw, IN, USA), povidone-iodine (Betadine), and chlorhexidine gluconate solution (Irrisept; Irrimax, Gainesville, FL, USA) in reducing biofilm formation of Staphylococcus aureus, Staphylococcus epidermidis, and Cutibacterium acnes inoculated on cobalt-chrome, titanium, and stainless steel disks, representing metals commonly used for shoulder arthroplasty. The hypothesis was that there would be no significant difference in biofilm reduction among the 3 topical adjuvants. METHODS Strains of S aureus (ATCC 35556), S epidermidis (ATCC 35984), and C acnes (LMG 16711) were grown on cobalt-chrome, titanium, and stainless steel disks. For each strain, the disks were divided into 4 groups: (1) control, (2) povidone-iodine (Betadine), (3) chlorhexidine gluconate (Irrisept), and (4) Bactisure. Bacteria were grown on 5% sheep blood agar plates. Biofilm eradication was quantified using adenosine triphosphate bioluminescence and compared with controls 48 and 72 hours after implementation of the topical adjuvant. RESULTS At 72 hours after implementation of the topical adjuvant, a statistically significant reduction in colony-forming units was observed for all topical adjuvants across all tested metals, as compared with their respective control. With respect to the topical adjuvants themselves, Bactisure more consistently demonstrated the most significant reduction in colony-forming units across all bacteria when the tested medium was adjusted for, with the exception of S aureus, which showed similar results to Betadine at 72 hours. CONCLUSION By use of commonly encountered topical adjuvants on S aureus-, S epidermidis-, and C acnes-inoculated disks of various implant metals, a significant reduction in biofilm production was observed. Bactisure, a recent Food and Drug Administration-approved topical adjuvant, demonstrated the overall greatest efficacy of the agents studied.
Collapse
Affiliation(s)
- Cameron Kia
- Department of Orthopedic Surgery, UConn Health, Farmington, CT, USA
| | - Antonio Cusano
- Department of Orthopedic Surgery, UConn Health, Farmington, CT, USA.
| | - James Messina
- Department of Orthopedic Surgery, UConn Health, Farmington, CT, USA
| | - Lukas N Muench
- Department of Orthopedic Surgery, UConn Health, Farmington, CT, USA; Department of Orthopaedic Sports Medicine, Technical University of Munich, Munich, Germany
| | | | | | | | | |
Collapse
|
78
|
Rozis M, Evangelopoulos DS, Pneumaticos SG. Orthopedic Implant-Related Biofilm Pathophysiology: A Review of the Literature. Cureus 2021; 13:e15634. [PMID: 34306846 PMCID: PMC8278357 DOI: 10.7759/cureus.15634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2021] [Indexed: 11/05/2022] Open
Abstract
Orthopedic implant-related infections remain a major problem even nowadays. Bacterial resistance through biofilm formation, in addition to the limited treatment options available, has resulted in an increased effort to better understand pathophysiology mechanisms. We performed a review of the literature in order to identify major biofilm formation pathways through which possible treatment strategies could arise.
Collapse
Affiliation(s)
- Meletis Rozis
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT Hospital, Athens, GRC
| | | | - Spyros G Pneumaticos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT Hospital, Athens, GRC
| |
Collapse
|
79
|
Skiba-Kurek I, Nowak P, Empel J, Tomczak M, Klepacka J, Sowa-Sierant I, Żak I, Pomierny B, Karczewska E. Evaluation of Biofilm Formation and Prevalence of Multidrug-Resistant Strains of Staphylococcus epidermidis Isolated from Neonates with Sepsis in Southern Poland. Pathogens 2021; 10:pathogens10070877. [PMID: 34358027 PMCID: PMC8308537 DOI: 10.3390/pathogens10070877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 01/11/2023] Open
Abstract
Staphylococcus epidermidis strains play an important role in nosocomial infections, especially in the ones associated with biofilm formation on medical devices. The paper was aimed at analyzing the mechanisms of antibiotic resistance and confirming the biofilm-forming ability among S. epidermidis strains isolated from the blood of hospitalized newborns. Genetic analysis of resistance mechanism determinants included multiplex PCR detection of mecA, ermA, ermB, ermC, msrA, and mef genes. Biofilm analysis comprised phenotypic and genotypic methods including Christensen and Freeman methods and PCR detection of the icaADB gene complex. Among the tested S. epidermidis strains, 89% of the isolates were resistant to methicillin, 67%—to erythromycin, 53%—to clindamycin, 63%—to gentamicin, and 23%—to teicoplanin, while all the strains were susceptible to vancomycin and linezolid. The mecA gene was detected in 89% of the isolates, the ermC gene was the most common and present among 56% of the strains, while the msrA gene was observed in 11% isolates. Eighty-five percent of the strains were described as biofilm-positive by phenotypic methods and carried the icaADB gene cluster. Multidrug resistance and the biofilm-forming ability in most of the strains tested may contribute to antimicrobial therapy failure (p < 0.05).
Collapse
Affiliation(s)
- Iwona Skiba-Kurek
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Krakow, Poland; (I.S.-K.); (P.N.)
| | - Paweł Nowak
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Krakow, Poland; (I.S.-K.); (P.N.)
| | - Joanna Empel
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, Chełmska 30/34 Street, 00-725 Warsaw, Poland; (J.E.); (M.T.)
| | - Magdalena Tomczak
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, Chełmska 30/34 Street, 00-725 Warsaw, Poland; (J.E.); (M.T.)
| | - Joanna Klepacka
- Department of Clinical Microbiology, University Children’s Hospital of Krakow, Wielicka 256 Street, 30-663 Krakow, Poland; (J.K.); (I.S.-S.); (I.Ż.)
| | - Iwona Sowa-Sierant
- Department of Clinical Microbiology, University Children’s Hospital of Krakow, Wielicka 256 Street, 30-663 Krakow, Poland; (J.K.); (I.S.-S.); (I.Ż.)
| | - Iwona Żak
- Department of Clinical Microbiology, University Children’s Hospital of Krakow, Wielicka 256 Street, 30-663 Krakow, Poland; (J.K.); (I.S.-S.); (I.Ż.)
| | - Bartosz Pomierny
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland;
| | - Elżbieta Karczewska
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Krakow, Poland; (I.S.-K.); (P.N.)
- Correspondence: ; Tel.: +481-2620-5750; Fax: +481-2620-5758
| |
Collapse
|
80
|
Nasser A, Dallal MMS, Jahanbakhshi S, Azimi T, Nikouei L. Staphylococcus aureus: biofilm formation and strategies against it. Curr Pharm Biotechnol 2021; 23:664-678. [PMID: 34238148 DOI: 10.2174/1389201022666210708171123] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/09/2021] [Accepted: 05/31/2021] [Indexed: 11/22/2022]
Abstract
The formation of Staphylococcus aureus biofilm causes significant infections in the human body. Biofilm forms through the aggregation of bacterial species and brings about many complications. It mediates drug resistance and persistence and facilitates the recurrence of infection at the end of antimicrobial therapy. Biofilm formation goes through a series of steps to complete, and any interference in these steps can disrupt its formation. Such interference may occur at any stage of biofilm production, including attachment, monolayer formation, and accumulation. Interfering agents can act as quorum sensing inhibitors and interfere in the functionality of quorum sensing receptors, attachment inhibitors and affect the cell hydrophobicity. Among these inhibiting strategies, attachment inhibitors could serve as the best agents against biofilm formation. If pathogens abort the attachment, the following stages of biofilm formation, e.g., accumulation and dispersion, will fail to materialize. Inhibition at this stage leads to suppression of virulence factors and invasion. One of the best-known inhibitors is a chelator that collects metal, Fe+, Zn+, and magnesium critical for biofilm formation. These influential factors in the binding and formation of biofilm are investigated, and the coping strategy is discussed. This review examines the stages of biofilm formation and determines what factors interfere in the continuity of these steps. Finally, the inhibition strategies are investigated, reviewed, and discussed. Keywords: Biofilm, Staphylococcus, Biofilm inhibitor, Dispersion, Antibiofilm agent, EPS, PIA.
Collapse
Affiliation(s)
- Ahmad Nasser
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shiva Jahanbakhshi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Taher Azimi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Nikouei
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
81
|
Photodynamic Therapy Combined with Antibiotics or Antifungals against Microorganisms That Cause Skin and Soft Tissue Infections: A Planktonic and Biofilm Approach to Overcome Resistances. Pharmaceuticals (Basel) 2021; 14:ph14070603. [PMID: 34201530 PMCID: PMC8308592 DOI: 10.3390/ph14070603] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022] Open
Abstract
The present review covers combination approaches of antimicrobial photodynamic therapy (aPDT) plus antibiotics or antifungals to attack bacteria and fungi in vitro (both planktonic and biofilm forms) focused on those microorganisms that cause infections in skin and soft tissues. The combination can prevent failure in the fight against these microorganisms: antimicrobial drugs can increase the susceptibility of microorganisms to aPDT and prevent the possibility of regrowth of those that were not inactivated during the irradiation; meanwhile, aPDT is effective regardless of the resistance pattern of the strain and their use does not contribute to the selection of antimicrobial resistance. Additive or synergistic antimicrobial effects in vitro are evaluated and the best combinations are presented. The use of combined treatment of aPDT with antimicrobials could help overcome the difficulty of fighting high level of resistance microorganisms and, as it is a multi-target approach, it could make the selection of resistant microorganisms more difficult.
Collapse
|
82
|
S. epidermidis Isolates from a Tertiary Care Portuguese Hospital Show Very High Antibiotic Non-Susceptible Rates and Significant Ability to Form Biofilms. Appl Microbiol 2021. [DOI: 10.3390/applmicrobiol1010012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Healthcare-associated infections (HAIs) have been increasing during recent decades, leading to long hospital stays and high morbidity and mortality rates. The usage of antibiotics therapy against these infections is enhancing the emergence of more multiple-drug resistant strains, in particular in Staphylococcus epidermidis. Hence, this study focused on the resistance pattern of S. epidermidis isolates from clinical settings and its association with phenotypic and molecular traits. Our results showed that HAIs were more prevalent among infants and older adults, and the most frequent type of HAI was central line-associated bloodstream infection. Half of the patients received antibiotic therapy before laboratory diagnosis. Preceding microbiological diagnosis, the number of patients receiving antibiotic therapy increased by 29.1%. Eighty-six per cent of the clinical isolates presented a multidrug resistance (MDR) profile, and a quarter were strong biofilm producers. Furthermore, polysaccharide intercellular adhesin (PIA)-dependent biofilms presented higher biomass production (p = 0.0041) and a higher rate of antibiotic non-susceptibility than PIA-independent biofilms, emphasizing the role of icaABDC operon in infection severity. Therefore, this study suggests that a thorough understanding of the phenotypic and molecular traits of the bacterial cause of the HAIs may lead to a more suitable selection of antibiotic therapy, improving guidance and outcome assessment.
Collapse
|
83
|
Loza-Correa M, Yousuf B, Ramirez-Arcos S. Staphylococcus epidermidis undergoes global changes in gene expression during biofilm maturation in platelet concentrates. Transfusion 2021; 61:2146-2158. [PMID: 33904608 DOI: 10.1111/trf.16418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Staphylococcus epidermidis forms surface-attached aggregates (biofilms) when grown in platelet concentrates (PCs). Comparative transcriptome analyses were undertaken to investigate differential gene expression of S. epidermidis biofilms grown in PCs. STUDY DESIGN AND METHODS Two S. epidermidis strains isolated from human skin (AZ22 and AZ39) and one strain isolated from contaminated PCs (ST02) were grown in glucose-supplemented Trypticase Soy Broth (TSBg) and PCs. RNA was extracted and sequenced using Illumina HiSeq. Differential expression analysis was done using DESeq, and significantly differentially expressed genes (DEGs) were selected. DEGs were subjected to Kyoto encyclopedia of genes and genomes and Gene Ontology analyses. Differential gene expression was validated with quantitative reverse transcription-PCR. RESULTS A total of 436, 442, and 384 genes were expressed in AZ22, AZ39, and ST02, respectively. DEG analysis showed that 170, 172, and 117 genes were upregulated in PCs in comparison to TSBg, whereas 120, 135, and 89 genes were downregulated (p < .05) in mature biofilms of AZ22, AZ39, and ST02, respectively. Twenty-seven DEGs were shared by all three strains. While 76 DEGs were shared by AZ22 and AZ39, only 34 and 21 DEGs were common between ST02, and AZ22 and AZ39, respectively. Significant transcriptional expression changes were observed in genes involved in platelet-bacteria interaction, biofilm formation, production of virulence factors, and resistance to antimicrobial peptides and antibiotics. CONCLUSION Differential gene expression in S. epidermidis is triggered by the stressful PC storage environment. Upregulation of virulence and antimicrobial resistance genes could have clinical implications for transfusion patients.
Collapse
Affiliation(s)
- Maria Loza-Correa
- Centre for Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Basit Yousuf
- Centre for Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Sandra Ramirez-Arcos
- Centre for Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
84
|
Boix-Lemonche G, Guillem-Marti J, Lekka M, D'Este F, Guida F, Manero JM, Skerlavaj B. Membrane perturbation, altered morphology and killing of Staphylococcus epidermidis upon contact with a cytocompatible peptide-based antibacterial surface. Colloids Surf B Biointerfaces 2021; 203:111745. [PMID: 33853003 DOI: 10.1016/j.colsurfb.2021.111745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/08/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022]
Abstract
One possibility to prevent prosthetic infections is to produce biomaterials resistant to bacterial colonization by anchoring membrane active antimicrobial peptides (AMPs) onto the implant surface. In this perspective, a deeper understanding of the mode of action of the immobilized peptides should improve the development of AMP-inspired infection-resistant biomaterials. The aim of the present study was to characterize the bactericidal mechanism against Staphylococcus epidermidis of the AMP BMAP27(1-18), immobilized on titanium disks and on a model resin support, by applying viability counts, Field Emission Scanning Electron Microscopy (FE-SEM), and a fluorescence microplate assay with a membrane potential-sensitive dye. The cytocompatibility to osteoblast-like MG-63 cells was investigated in monoculture and in co-culture with bacteria. The impact of peptide orientation was explored by using N- and C- anchored analogues. On titanium, the ∼50 % drop in bacteria viability and dramatically affected morphology indicate a contact-killing action exerted by the N- and C-immobilized peptides to the same extent. As further shown by the fluorescence assay with the resin-anchored peptides, the bactericidal effect was mediated by rapid membrane perturbation, similar to free peptides. However, at peptide MBC resin equivalents the C-oriented analogue proved more effective with more than 99 % killing and maximum fluorescence increase, compared to half-maximum fluorescence with more than 90 % killing produced by the N-orientation. Confocal microscopy analyses revealed 4-5 times better MG-63 cell adhesion on peptide-functionalized titanium both in monoculture and in co-culture with bacteria, regardless of peptide orientation, thus stimulating further studies on the effects of the immobilized BMAP27(1-18) on osteoblast cells.
Collapse
Affiliation(s)
- Gerard Boix-Lemonche
- Department of Medicine (DAME), University of Udine, piazzale Kolbe, 4, 33100, Udine, Italy.
| | - Jordi Guillem-Marti
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 14, 08930 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering-UPC, Av. Eduard Maristany 14, 08930, Barcelona, Spain.
| | - Maria Lekka
- University of Udine, Polytechnic Department of Engineering and Architecture, Via delle Scienze 206, 33100, Udine, Italy; CIDETEC, Basque Research and Technology Alliance (BRTA), Po. Miramón 196, 20014 Donostia-San Sebastián, Spain.
| | - Francesca D'Este
- Department of Medicine (DAME), University of Udine, piazzale Kolbe, 4, 33100, Udine, Italy.
| | - Filomena Guida
- University of Trieste, Department of Life Sciences, Via Giorgieri 5, 34127, Trieste, Italy.
| | - José María Manero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 14, 08930 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering-UPC, Av. Eduard Maristany 14, 08930, Barcelona, Spain.
| | - Barbara Skerlavaj
- Department of Medicine (DAME), University of Udine, piazzale Kolbe, 4, 33100, Udine, Italy.
| |
Collapse
|
85
|
Marincola G, Jaschkowitz G, Kieninger AK, Wencker FDR, Feßler AT, Schwarz S, Ziebuhr W. Plasmid-Chromosome Crosstalk in Staphylococcus aureus: A Horizontally Acquired Transcription Regulator Controls Polysaccharide Intercellular Adhesin-Mediated Biofilm Formation. Front Cell Infect Microbiol 2021; 11:660702. [PMID: 33829001 PMCID: PMC8019970 DOI: 10.3389/fcimb.2021.660702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/02/2021] [Indexed: 11/24/2022] Open
Abstract
Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) of clonal complex CC398 typically carry various antimicrobial resistance genes, many of them located on plasmids. In the bovine LA-MRSA isolate Rd11, we previously identified plasmid pAFS11 in which resistance genes are co-localized with a novel ica-like gene cluster, harboring genes required for polysaccharide intercellular adhesin (PIA)-mediated biofilm formation. The ica genes on pAFS11 were acquired in addition to a pre-existing ica locus on the S. aureus Rd11 chromosomal DNA. Both loci consist of an icaADBC operon and icaR, encoding a corresponding icaADBC repressor. Despite carrying two biofilm gene copies, strain Rd11 did not produce PIA and transformation of pAFS11 into another S. aureus strain even slightly diminished PIA-mediated biofilm formation. By focusing on the molecular background of the biofilm-negative phenotype of pAFS11-carrying S. aureus, we identified the pAFS11-borne ica locus copy as functionally fully active. However, transcription of both plasmid- and core genome-derived icaADBC operons were efficiently suppressed involving IcaR. Surprisingly, although being different on the amino acid sequence level, the two IcaR repressor proteins are mutually replaceable and are able to interact with the icaA promoter region of the other copy. We speculate that this regulatory crosstalk causes the biofilm-negative phenotype in S. aureus Rd11. The data shed light on an unexpected regulatory interplay between pre-existing and newly acquired DNA traits in S. aureus. This also raises interesting general questions regarding functional consequences of gene transfer events and their putative implications for the adaptation and evolution of bacterial pathogens.
Collapse
Affiliation(s)
- Gabriella Marincola
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Greta Jaschkowitz
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Ann-Katrin Kieninger
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Freya D R Wencker
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Andrea T Feßler
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Free University of Berlin, Berlin, Germany
| | - Stefan Schwarz
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Free University of Berlin, Berlin, Germany
| | - Wilma Ziebuhr
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
86
|
Nguyen PT, Nguyen MT, Bolhuis A. Inhibition of biofilm formation by alpha-mangostin loaded nanoparticles against Staphylococcus aureus. Saudi J Biol Sci 2021; 28:1615-1621. [PMID: 33732047 PMCID: PMC7938154 DOI: 10.1016/j.sjbs.2020.11.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/14/2020] [Indexed: 11/17/2022] Open
Abstract
This study aimed to investigate the antibiofilm activity of alpha-mangostin (AMG) loaded nanoparticles (nanoAMG) against Staphylococcus aureus, including the methicillin-resistant strain MRSA252. The results indicated that treatment with 24 μmol/L nanoAMG inhibited the formation of biofilm biomass by 53-62%, compared to 40-44% for free AMG (p < 0.05). At 48 μmol/L, biofilms in all nanoAMG treated samples were nearly fully disrupted for the two tested strains, MRSA252 and the methicillin-sensitive strain NCTC6571. That concentration resulted in killing of biofilm cells. A lower concentration of 12 µmol/L nanoAMG inhibited initial adherence of the two bacterial strains by > 50%. In contrast, activity of nanoAMG was limited on preformed mature biofilms, which at a concentration of 48 µmol/L were reduced only by 27% and 22% for NCTC6571 and MRSA252, respectively. The effects of AMG or nanoAMG on the expression of biofilm-related genes showed some noticeable differences between the two strains. For instance, the expression level of ebpS was downregulated in MRSA252 and upregulated in NCTC6571 when those strains were treated with either AMG or nanoAMG. In contrast, the expression of fnbB was down regulated in NCTC6571, while it was up-regulated in the MRSA252. The expression of other biofilm-related genes (icaC, clfB and fnbA) was down regulated in both strains. In conclusion, our results suggest that AMG coated nanoparticles had enhanced biological activity as compared to free AMG, indicating that nanoAMG could be a new and promising inhibitor of biofilm formation to tackle S. aureus, including strains that are resistant to multiple antibiotics.
Collapse
Affiliation(s)
- Phuong T.M. Nguyen
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay, Hanoi, Viet Nam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay, Hanoi, Viet Nam
- Corresponding authors at: Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay, Hanoi, Viet Nam.
| | - Minh T.H. Nguyen
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Road, Cau Giay, Hanoi, Viet Nam
| | - Albert Bolhuis
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK
- Corresponding authors at: Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay, Hanoi, Viet Nam.
| |
Collapse
|
87
|
Hutchings C, Prokocimer Yair Z, Reifen R, Shemesh M. Antimicrobial Effect of Zn 2+ Ions Governs the Microbial Quality of Donor Human Milk. Foods 2021; 10:637. [PMID: 33802996 PMCID: PMC8002753 DOI: 10.3390/foods10030637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 11/21/2022] Open
Abstract
Donor human milk (HM) obtained at HM banks is exceptionally crucial for the feeding and treatment of preterm infants. Bacterial contaminations of HM in various stages of its handling are very common and can lead to disqualification of donations or severe infections in worse cases. Hence, HM donations are subject to strict bacteriological evaluations pre- and post-pasteurization. The main contaminating species vary between countries, banks and donors and even exhibit inter-individual variation. We initiated an assessment of the bacteriological composition of HM donated by women hospitalized in a neonatal intensive care unit in Israel. The most common bacterium identified was Staphylococcus epidermidis, found in all but one of the HM samples; the presence of several species of coagulase-negative Staphylococci was also noted. Next, we sought to develop a platform towards antibacterial treatment using Zn2+ ions that have recently been found to be potent against contaminants isolated from bovine milk. Zn2+ efficiently inhibited the growth of viable aerobic population and S. epidermidis in HM. Growth was also inhibited in other Gram-positive bacteria such as Bacillus cereus, a well-known food-borne pathogen. S. epidermidis and B. cereus cells grown in the presence of zinc were taken for microscopic evaluation, aiming to demonstrate zinc's antimicrobial mode of action morphologically. Images obtained using scanning electron microscopy indicated leakage of cellular content and cell lysis in S. epidermidis. Besides, B. cereus cells showed abnormalities in their cell surface and complete loss of flagella upon treatment with zinc. Along with the above findings, it should be noted that this was a pilot study that tested how high doses of Zn2+ affect breast milk as a product. Further research is likely needed on the safety of consumption of Zn2+-treated HM in infants and older children.
Collapse
Affiliation(s)
- Carmel Hutchings
- Department of Food Sciences, Institute for Postharvest Technology and Food Sciences, Agricultural Research Organization (ARO), Volcani Center, Rishon LeZion 7528809, Israel;
- School of Nutrition Science, Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot 761001, Israel
| | - Zafnat Prokocimer Yair
- Schneider Children’s Medical Center Israel, Emergency Medicine Department, Petah-Tikva 49202, Israel;
| | - Ram Reifen
- School of Nutrition Science, Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot 761001, Israel
| | - Moshe Shemesh
- Department of Food Sciences, Institute for Postharvest Technology and Food Sciences, Agricultural Research Organization (ARO), Volcani Center, Rishon LeZion 7528809, Israel;
| |
Collapse
|
88
|
Complete Genome Sequence of Staphylococcus epidermidis PH1-28, Isolated from the Forehead of a Hyperseborrheic Donor. Microbiol Resour Announc 2021; 10:10/9/e00165-21. [PMID: 33664152 PMCID: PMC7936632 DOI: 10.1128/mra.00165-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We report the complete genome sequence of Staphylococcus epidermidis commensal strain PH1-28, isolated from the forehead of a healthy donor. The assembled 2.6-Mbp genome consisted of one chromosome and five plasmids. These data will provide valuable information and important insights into the physiology and metabolism of this skin flora microorganism.
Collapse
|
89
|
Wark KJL, Cains GD. The Microbiome in Hidradenitis Suppurativa: A Review. Dermatol Ther (Heidelb) 2021; 11:39-52. [PMID: 33244661 PMCID: PMC7859000 DOI: 10.1007/s13555-020-00465-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Hidradenitis suppurativa (HS) is a chronic autoinflammatory skin disease. It is characterised by the development of abscesses and nodules in intertriginous anatomical sites. Whilst it is now recognised as an autoinflammatory condition rather than an infective disease, bacteria are implicated in disease pathogenesis. METHODS We performed a search of the literature from inception to 12 August 2020 using the search terms "hidradenitis suppurativa", "Verneuil's disease", "acne inversa", "microbiome", "bacteriology" and "microbiology". Studies were included if they assessed the cutaneous, gut or oral bacteria, bacteriology or microbiome in hidradenitis suppurativa. RESULTS Twenty-one studies examining the cutaneous microbiome and two studies examining the gastrointestinal microbiome in HS were identified. No studies examining the oral microbiome in HS were identified. A total of 972 patients and 46 healthy controls were included across studies examining the cutaneous microbiome. A total of 100 patients and 36 controls were included across both gut microbiome studies. Coagulase-negative Staphylococcus, anaerobes such as Porphyromonas and Prevotella, and Staphylococcus aureus species were commonly encountered organisms across the included cutaneous microbiome studies. The studies examining the gut microbiome were limited, with one small study demonstrating an alteration in the gut microbiome composition compared to controls. The other study found no alteration to the gut microbiome in patients with HS compared to those with inflammatory bowel disease (IBD) and HS, and IBD and/or psoriasis. CONCLUSION Research should be undertaken into the oral microbiome in HS. Further research should be undertaken examining the cutaneous and gut microbiome in HS, and its relationship with documented co-morbidities. Additionally, metagenomics-focused studies may help identify the relationship between microorganisms and host, and this may shed light on new pathways of disease pathogenesis. This may help identify potential future therapeutic targets.
Collapse
Affiliation(s)
- Kirsty J L Wark
- Department of Dermatology, Liverpool Hospital, Sydney, Australia.
- School of Medicine, University of New South Wales, Sydney, Australia.
| | - Geoffrey D Cains
- Department of Dermatology, Liverpool Hospital, Sydney, Australia
- School of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
90
|
Inhibition of Biofilm Formation by the Synergistic Action of EGCG-S and Antibiotics. Antibiotics (Basel) 2021; 10:antibiotics10020102. [PMID: 33494273 PMCID: PMC7909832 DOI: 10.3390/antibiotics10020102] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/18/2022] Open
Abstract
Biofilm, a stress-induced physiological state, is an established means of antimicrobial tolerance. A perpetual increase in multidrug resistant (MDR) infections associated with high mortality and morbidity have been observed in healthcare settings. Multiple studies have indicated that the use of natural products can prevent bacterial growth. Recent studies in the field have identified that epigallocatechin gallate (EGCG), a green tea polyphenol, could disrupt bacterial biofilms. A modified lipid-soluble EGCG, epigallocatechin-3-gallate-stearate (EGCG-S), has enhanced the beneficial properties of green tea. This study focuses on utilizing EGCG-S as a novel synergistic agent with antibiotics to prevent or control biofilm. Different formulations of EGCG-S and selected antibiotics were used to study their combinatorial effects on biofilms produced by five potential pathogenic bacteria, Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, Staphylococcus epidermidis, and Mycobacterium smegmatis. The crystal violet (CV) assay and the sensitive fluorescence-based resazurin biofilm viability assay were used to assess the biofilm production. Our results identified optimal formulation for each bacterium, effectively inhibiting biofilm formation to an extent of 95-99%. Colony-forming unit (CFU) and cell viability analyses showed a decrease of viable bacteria. These results depict the potential of EGCG-S as a synergistic agent with antibiotics and as an anti-biofilm agent.
Collapse
|
91
|
Pelletier J, Ocean Ophthalmology Group, Miami, FL, USA. The Role of Biofilms in Pathology of the Ocular Surface. Ophthalmology 2021. [DOI: 10.17925/opht.2021.15.1.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
92
|
Abstract
Plants that are primarily used as a food source commonly have undervalued biological properties beyond the basic supply of nutrients. One important example of this are the antimicrobial properties of plants. Inclusion of natural and food grade antimicrobial ingredients in recipes to prevent food spoilage and disease transmission, or in cosmetic products to prevent transient and pathogenic bacteria would have world-wide public health implications. A patented natural polyphenol rich sugar cane extract (PRSE), is marketed as a high anti-oxidant and polyphenol ingredient, but its anti-microbial activity has not been reported previously. We determined the anti-bacterial properties of PRSE on common human pathogens relating to a range of diseases including food poisoning, tooth decay, acne and severe skin infections using disc/well diffusion experiments. Our findings indicate that PRSE is an efficient antimicrobial, which could be included at differing dosages to target a range of food borne and environmental pathogens.
Collapse
|
93
|
Cataneli Pereira V, Pinheiro-Hubinger L, de Oliveira A, Moraes Riboli DF, Benini Martins K, Calixto Romero L, Ribeiro de Souza da Cunha MDL. Detection of the agr System and Resistance to Antimicrobials in Biofilm-Producing S. epidermidis. Molecules 2020; 25:molecules25235715. [PMID: 33287389 PMCID: PMC7729762 DOI: 10.3390/molecules25235715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 11/19/2022] Open
Abstract
The ability of Staphylococcus epidermidis to produce virulence factors, such as biofilm, added to its increased resistance to antimicrobials can cause infections that are difficult to treat. Many staphylococcal virulence factors are under the control of the accessory gene regulator (agr). The objective of this study was to establish the agr locus and susceptibility of biofilm-producing S. epidermidis specimens to antimicrobial agents, through PCR reactions, reverse transcription polymerase chain reaction (RT-PCR), and the determination of minimum inhibitory concentration (MIC), and to analyze the clonal profile of 300 strains isolated from blood culture specimens from inpatients at a University Hospital in Brazil, over a 20-year period by pulsed-field gel electrophoresis (PFGE) and multilocus sequence typing (MLST) techniques. The ica operon expression was shown in 83.6% strains, bhp gene in 11.5%, and aap gene in 32.8%. Oxacillin resistance was detected in 90.1%, while 4.9% showed tigecycline resistance, and intermediate resistance to quinupristin/dalfopristin was identified in 0.4%. Clonal profile determination showed 11 clusters, with the ST2 type determined as the major cluster. The S. epidermidis biofilm producer demonstrated a predominance of agr I locus, oxacillin resistance, and SCCmec III as well as the potential dissemination of pathogenic clones in hospital settings over long periods.
Collapse
Affiliation(s)
- Valéria Cataneli Pereira
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
- Sector of Microbiology and Immunology, UNOESTE—University of West Paulista, Presidente Prudente CEP 19050-920, São Paulo, Brazil
| | - Luiza Pinheiro-Hubinger
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Adilson de Oliveira
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Danilo Flávio Moraes Riboli
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Katheryne Benini Martins
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Letícia Calixto Romero
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
| | - Maria de Lourdes Ribeiro de Souza da Cunha
- Sector of Microbiology and Immunology, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP—University Estadual Paulista, Botucatu CEP 18618-689, São Paulo, Brazil; (V.C.P.); (L.P.-H.); (A.d.O.); (D.F.M.R.); (K.B.M.); (L.C.R.)
- Correspondence: ; Tel.: +55-14-3880-0428
| |
Collapse
|
94
|
Fournière M, Latire T, Souak D, Feuilloley MGJ, Bedoux G. Staphylococcus epidermidis and Cutibacterium acnes: Two Major Sentinels of Skin Microbiota and the Influence of Cosmetics. Microorganisms 2020; 8:E1752. [PMID: 33171837 PMCID: PMC7695133 DOI: 10.3390/microorganisms8111752] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/26/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Dermatological and cosmetics fields have recently started to focus on the human skin microbiome and microbiota, since the skin microbiota is involved in the health and dysbiosis of the skin ecosystem. Amongst the skin microorganisms, Staphylococcus epidermidis and Cutibacterium acnes, both commensal bacteria, appear as skin microbiota sentinels. These sentinels have a key role in the skin ecosystem since they protect and prevent microbiota disequilibrium by fighting pathogens and participate in skin homeostasis through the production of beneficial bacterial metabolites. These bacteria adapt to changing skin microenvironments and can shift to being opportunistic pathogens, forming biofilms, and thus are involved in common skin dysbiosis, such as acne or atopic dermatitis. The current evaluation methods for cosmetic active ingredient development are discussed targeting these two sentinels with their assets and limits. After identification of these objectives, research of the active cosmetic ingredients and products that maintain and promote these commensal metabolisms, or reduce their pathogenic forms, are now the new challenges of the skincare industry in correlation with the constant development of adapted evaluation methods.
Collapse
Affiliation(s)
- Mathilde Fournière
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (T.L.); (G.B.)
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Catholique de l’Ouest Bretagne Nord, 22200 Guingamp, France
| | - Thomas Latire
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (T.L.); (G.B.)
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Catholique de l’Ouest Bretagne Nord, 22200 Guingamp, France
| | - Djouhar Souak
- Laboratoire de Microbiologie Signaux et Microenvironment LMSM EA4312, Université de Rouen Normandie, 27000 Évreux, France; (D.S.); (M.G.J.F.)
- BASF Beauty Care Solutions France SAS, 69007 Lyon, France
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironment LMSM EA4312, Université de Rouen Normandie, 27000 Évreux, France; (D.S.); (M.G.J.F.)
| | - Gilles Bedoux
- Laboratoire de Biotechnologie et Chimie Marines LBCM EA 3884, IUEM, Université Bretagne Sud, 56000 Vannes, France; (T.L.); (G.B.)
| |
Collapse
|
95
|
Bottagisio M, Barbacini P, Bidossi A, Torretta E, deLancey-Pulcini E, Gelfi C, James GA, Lovati AB, Capitanio D. Phenotypic Modulation of Biofilm Formation in a Staphylococcus epidermidis Orthopedic Clinical Isolate Grown Under Different Mechanical Stimuli: Contribution From a Combined Proteomic Study. Front Microbiol 2020; 11:565914. [PMID: 33013797 PMCID: PMC7505995 DOI: 10.3389/fmicb.2020.565914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/18/2020] [Indexed: 11/13/2022] Open
Abstract
One of the major causes of prosthetic joint failure is infection. Recently, coagulase negative Staphylococcus epidermidis has been identified as an emergent, nosocomial pathogen involved in subclinical prosthetic joint infections (PJIs). The diagnosis of PJIs mediated by S. epidermidis is usually complex and difficult due to the absence of acute clinical signs derived from the host immune system response. Therefore, analysis of protein patterns in biofilm-producing S. epidermidis allows for the examination of the molecular basis of biofilm formation. Thus, in the present study, the proteome of a clinical isolate S. epidermidis was analyzed when cultured in its planktonic or sessile form to examine protein expression changes depending on culture conditions. After 24 h of culture, sessile bacteria exhibited increased gene expression for ribosomal activity and for production of proteins related to the initial attachment phase, involved in the capsular polysaccharide/adhesin, surface associated proteins and peptidoglycan biosynthesis. Likewise, planktonic S. epidermidis was able to aggregate after 24 h, synthesizing the accumulation associate protein and cell-wall molecules through the activation of the YycFG and ArlRS, two component regulatory pathways. Prolonged culture under vigorous agitation generated a stressful growing environment triggering aggregation in a biofilm-like matrix as a mechanism to survive harsh conditions. Further studies will be essential to support these findings in order to further delineate the complex mechanisms of biofilm formation of S. epidermidis and they could provide the groundwork for the development of new drugs against biofilm-related infections, as well as the identification of novel biomarkers of subclinical or chronic infections mediated by these emerging, low virulence pathogens.
Collapse
Affiliation(s)
- Marta Bottagisio
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Clinical Chemistry and Microbiology, Milan, Italy
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Alessandro Bidossi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Clinical Chemistry and Microbiology, Milan, Italy
| | | | - Elinor deLancey-Pulcini
- Medical Biofilm Laboratory, Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Garth A James
- Medical Biofilm Laboratory, Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States
| | - Arianna B Lovati
- IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, Milan, Italy
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
96
|
Ramesh S, Kovelakuntla V, Meyer AS, Rivero IV. Three-dimensional printing of stimuli-responsive hydrogel with antibacterial activity. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
97
|
Dutta K, Karmakar A, Jana D, Ballav S, Shityakov S, Panda AK, Ghosh C. Benzyl isocyanate isolated from the leaves of Psidium guajava inhibits Staphylococcus aureus biofilm formation. BIOFOULING 2020; 36:1000-1017. [PMID: 33172298 DOI: 10.1080/08927014.2020.1842877] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 09/03/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
Benzyl isocyanate (BIC), from methanol extract of Psidium guajava leaves, exhibited substantial anti-biofilm activities against Staphylococcus aureus, the common bacterial pathogen in nosocomial infections. Major components of the extract included eugenol, BIC, phenyl-2-methoxy-4-(1-propenyl)-acetate and 2,5-pyrrolidinedione,1-penta-3-4-dienyl, analyzed by GC-MS and HPLC studies. BIC exhibited substantial anti-biofilm activitiy against S. aureus, established by assaying biofilm formation, biofilm metabolic activity, bacterial adherence to hydrocarbons, exopolysaccharide formation, and optical and scanning electron microscopic studies. BIC significantly downregulated the important biofilm markers of S. aureus, viz., icaAD, sarA and agr, observed by quantitative real time polymerase chain reaction analysis. Molecular docking studies revealed thermodynamically favorable interaction of BIC with IcaA, SarA and Agr, having Gibbs energy values of -8.45, -9.09 and -10.29 kcal mol-1, respectively. BIC after binding to IcaR, the repressor of IcaA, influences its binding to target DNA site (Eshape, -157.27 kcal mol-1). The results are considered to demonstrate anti-biofilm potential of BIC against bacterial infections.
Collapse
Affiliation(s)
- Kunal Dutta
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Amit Karmakar
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Debarati Jana
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Saroj Ballav
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Sergey Shityakov
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Amiya Kumar Panda
- Department of Chemistry, Vidyasagar University, Midnapore, West Bengal, India
| | - Chandradipa Ghosh
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| |
Collapse
|
98
|
Lyu Z, Shang Y, Wang X, Wu Y, Zheng J, Liu H, Gong T, Ye L, Qu D. Monoclonal Antibodies Specific to the Extracellular Domain of Histidine Kinase YycG of Staphylococcus epidermidis Inhibit Biofilm Formation. Front Microbiol 2020; 11:1839. [PMID: 32849437 PMCID: PMC7426370 DOI: 10.3389/fmicb.2020.01839] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/14/2020] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus epidermidis is frequently associated with biofilm-related infections. Biofilms drastically reduce the efficacy of conventional antibiotics and the host immune system. In S. epidermidis biofilm formation, a major role is played by the YycG/YycF two-component system, and previous findings have indicated that inhibitors targeting the cytoplasmic HATPase_c domain of YycG kinase in S. epidermidis exhibit bactericidal and biofilm-killing activities. Therefore, we hypothesized that monoclonal antibodies (mAbs) against YycG extracellular (YycGex) domain would block the signal transduction and influence the biofilm formation of S. epidermidis. In this study, we screened out two YycGex-specific mAbs showing the highest affinity for the target, mAbs 2F3 and 1H1. These mAbs inhibited S. epidermidis biofilm formation in a dose-dependent manner, and at a concentration of 160 μg/mL, mAbs 2F3 and 1H1 caused 78.3 and 93.1% biofilm reduction, respectively, relative to normal mouse IgG control. When co-cultivated with YycGex mAbs, S. epidermidis cells showed diminished initial-adherence capacity, and the antibody treatment further led to a marked decrease in the synthesis of polysaccharide intercellular adhesin and in the transcriptional level of genes encoding proteins involved in biofilm formation. Lastly, we determined that the epitopes recognized by the two YycGex mAbs are located within aa 59–70 of the YycGex domain. It indicates that the YycGex domain may be a potential candidate as a vaccine for the prevention of S. epidermidis biofilm infections.
Collapse
Affiliation(s)
- Zhihui Lyu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongpeng Shang
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaofei Wang
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jinxin Zheng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital, Shenzhen University, Shenzhen, China
| | - Huayong Liu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ting Gong
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lina Ye
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
99
|
Cicciarella Modica D, Maurici M, D’Alò GL, Mozzetti C, Messina A, Distefano A, Pica F, De Filippis P. Taking Screenshots of the Invisible: A Study on Bacterial Contamination of Mobile Phones from University Students of Healthcare Professions in Rome, Italy. Microorganisms 2020; 8:microorganisms8071075. [PMID: 32707676 PMCID: PMC7409191 DOI: 10.3390/microorganisms8071075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/07/2023] Open
Abstract
Mobile phones (MPs) are commonly used both in the personal and professional life. We assessed microbiological contamination of MPs from 108 students in healthcare professions (HPs), in relation to their demographic characteristics and MPs handling habits, collected by means of a questionnaire. Cultural and biochemical tests were performed, and statistical analyses were carried out. Staphylococci were present in 85% of MPs, Enterococci in 37%, Coliforms in 6.5%; E. coli was never detected. Staphylococcus epidermidis was the most frequently isolated staphylococcal species (72% of MPs), followed by S. capitis (14%), S. saprophyticus, S. warneri, S. xylosus (6%), and by S. aureus (4%). Heterotrophic Plate Counts (HPC) at 37 °C, ranged from 0 to 1.2 × 104 CFU/dm2 (mean = 362 CFU/dm2). In univariate analysis, the male gender only was significantly associated with higher HPCs and enterococcal contamination. Multiple linear regression models explained only 17% and 16% of the HPC 37 °C and staphylococcal load variability, respectively. Developing specific guidelines for a hygienic use of MPs in clinical settings, for preventing cross-infection risks, is advisable, as well as introducing specific training programs to HP students. MPs decontamination procedures could also be implemented in the community.
Collapse
Affiliation(s)
| | - Massimo Maurici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (C.M.); (A.M.); (A.D.); (P.D.F.)
- Correspondence: (M.M.); (G.L.D.); Tel.: +39-327-8218-514 (G.L.D.)
| | - Gian Loreto D’Alò
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (C.M.); (A.M.); (A.D.); (P.D.F.)
- Department of Epidemiology, Lazio Regional Health Service, 00154 Rome, Italy
- Correspondence: (M.M.); (G.L.D.); Tel.: +39-327-8218-514 (G.L.D.)
| | - Cinzia Mozzetti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (C.M.); (A.M.); (A.D.); (P.D.F.)
| | - Alessandra Messina
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (C.M.); (A.M.); (A.D.); (P.D.F.)
| | - Alessandra Distefano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (C.M.); (A.M.); (A.D.); (P.D.F.)
| | - Francesca Pica
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Patrizia De Filippis
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (C.M.); (A.M.); (A.D.); (P.D.F.)
| |
Collapse
|
100
|
Zhou T, Wang XY, Deng DQ, Xu LH, Li XL, Guo Y, Li WH, Xie H, Zhang PL, Zhou XH. Nocardia colli sp. nov., a new pathogen isolated from a patient with primary cutaneous nocardiosis. Int J Syst Evol Microbiol 2020; 70:2981-2987. [PMID: 32375925 PMCID: PMC7395626 DOI: 10.1099/ijsem.0.003856] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
A novel nocardioform strain, CICC 11023T, was isolated from a tissue biopsy of neck lesions of a patient with primary cutaneous nocardiosis and characterized to establish its taxonomic position. The morphological, biochemical, physiological and chemotaxonomic properties of strain CICC 11023T were consistent with classification in the genus Nocardia. Whole-cell hydrolysates were rich in meso-diaminopimelic acid, galactose, arabinose and fructose. Mycolic acids were present. The major polar lipids were diphosphatidylglycerol, phosphatidylethanolamine, one unidentified phospholipid and two unidentified lipids, and the predominant menaquinone was cyclo MK-8 (H4, ω-cyclo). The main fatty acids (>5 %) were C18 : 0 10-methyl (TBSA), C16 : 0, summed feature 4 (C16 : 1 trans 9/C15 : 0 iso 2OH), C15 : 0 and C17 : 0 10-methyl. Phylogenetic analyses based on 16S rRNA gene sequences revealed that the isolate is most closely related (>98 % similarity) to the type strains Nocardia ninae OFN 02.72T, Nocardia iowensis UI 122540T and Nocardia alba YIM 30243T, and phylogenetic analysis of gyrB gene sequences showed similarity (89.1–92.2 %) to Nocardia vulneris NBRC 108936T, Nocardia brasiliensis IFM 0236T and Nocardia exalbida IFM 0803T. DNA–DNA hybridization results for strain CICC 11023T compared to Nocardia type strains ranged from 20.4 to 35.4 %. The genome of strain CICC 11023T was 8.78 Mbp with a G+C content of 67.4 mol% overall. The average nucleotide identity (ANI) values between strain CICC 11023T and N. alba YIM 30243T were low (OrthoANIu=77.47 %), and the ANI values between strain CICC 11023T and N. vulneris NBRC 108936 T were low (OrthoANIu=83.75 %). Consequently, strain CICC 11023T represents a novel Nocardia species on the basis of this polyphasic study, for which the name Nocardia colli sp. nov. is proposed. The type strain is CICC 11023T (=KCTC 39837T).
Collapse
Affiliation(s)
- Tao Zhou
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Xiao-Yun Wang
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
- *Correspondence: Xiao-Yun Wang,
| | - Dan-Qi Deng
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Li-Hua Xu
- The Key Laboratory of Microbial Diversity in Southwest China, Ministry of Education and Laboratory for Conservation and Utilization of Bio-resources, Yunnan Institute of Microbiology, Yunnan University, Kunming, Yunnan 650091, PR China
| | - Xiao-Lan Li
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Yun Guo
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Wen-Hua Li
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Hong Xie
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Pei-Lian Zhang
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Xiao-Hong Zhou
- Department of Dermatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| |
Collapse
|