1
|
Cavallo I, Sivori F, Mastrofrancesco A, Abril E, Pontone M, Di Domenico EG, Pimpinelli F. Bacterial Biofilm in Chronic Wounds and Possible Therapeutic Approaches. BIOLOGY 2024; 13:109. [PMID: 38392327 PMCID: PMC10886835 DOI: 10.3390/biology13020109] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
Wound repair and skin regeneration is a very complex orchestrated process that is generally composed of four phases: hemostasis, inflammation, proliferation, and remodeling. Each phase involves the activation of different cells and the production of various cytokines, chemokines, and other inflammatory mediators affecting the immune response. The microbial skin composition plays an important role in wound healing. Indeed, skin commensals are essential in the maintenance of the epidermal barrier function, regulation of the host immune response, and protection from invading pathogenic microorganisms. Chronic wounds are common and are considered a major public health problem due to their difficult-to-treat features and their frequent association with challenging chronic infections. These infections can be very tough to manage due to the ability of some bacteria to produce multicellular structures encapsulated into a matrix called biofilms. The bacterial species contained in the biofilm are often different, as is their capability to influence the healing of chronic wounds. Biofilms are, in fact, often tolerant and resistant to antibiotics and antiseptics, leading to the failure of treatment. For these reasons, biofilms impede appropriate treatment and, consequently, prolong the wound healing period. Hence, there is an urgent necessity to deepen the knowledge of the pathophysiology of delayed wound healing and to develop more effective therapeutic approaches able to restore tissue damage. This work covers the wound-healing process and the pathogenesis of chronic wounds infected by biofilm-forming pathogens. An overview of the strategies to counteract biofilm formation or to destroy existing biofilms is also provided.
Collapse
Affiliation(s)
- Ilaria Cavallo
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Francesca Sivori
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Arianna Mastrofrancesco
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Elva Abril
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Martina Pontone
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Enea Gino Di Domenico
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Fulvia Pimpinelli
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| |
Collapse
|
2
|
Shaghayegh G, Cooksley C, Bouras G, Houtak G, Nepal R, Psaltis AJ, Wormald PJ, Vreugde S. S. aureus biofilm metabolic activity correlates positively with patients' eosinophil frequencies and disease severity in chronic rhinosinusitis. Microbes Infect 2023; 25:105213. [PMID: 37652259 DOI: 10.1016/j.micinf.2023.105213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Chronic rhinosinusitis (CRS) is a persistent inflammation of the sinus mucosa. Recalcitrant CRS patients are unresponsive to medical and surgical interventions and often present with nasal polyps, tissue eosinophilia, and Staphylococcus aureus dominant mucosal biofilms. However, S. aureus sinonasal mucosal colonisation occurs in the absence of inflammation, questioning the role of S. aureus in CRS pathogenesis. Here, we aimed to investigate the relationship between S. aureus biofilm metabolic activity and virulence genes, innate immune cells, and disease severity in CRS. Biospecimens, including sinonasal tissue and nasal swabs, and clinical datasets, including disease severity scores, were obtained from CRS patients and non-CRS controls. S. aureus isolates were grown into biofilms in vitro, characterised, and sequenced. The patients' innate immune response was evaluated using flow cytometry. S. aureus was isolated in 6/19 (31.58%) controls and 23/53 (43.40%) CRS patients of 72 recruited patients. We found increased S. aureus biofilm metabolic activity in relation to increased eosinophil cell frequencies and disease severity in recalcitrant CRS cases. Mast cell frequencies were higher in tissue samples of patients carrying S. aureus harbouring lukF.PV, sea, and fnbB genes. Patients with S. aureus harbouring lukF.PV and sdrE genes had more severe disease. This offers insights into the pathophysiology of CRS and could lead to the development of more targeted therapies.
Collapse
Affiliation(s)
- Gohar Shaghayegh
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Clare Cooksley
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - George Bouras
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Ghais Houtak
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Roshan Nepal
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Alkis James Psaltis
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Peter-John Wormald
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Sarah Vreugde
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia.
| |
Collapse
|
3
|
Patyra E, Kwiatek K. Insect Meals and Insect Antimicrobial Peptides as an Alternative for Antibiotics and Growth Promoters in Livestock Production. Pathogens 2023; 12:854. [PMID: 37375544 DOI: 10.3390/pathogens12060854] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The extensive use of antibiotics in animal production has led to the development of antibiotic-resistant microorganisms and the search for alternative antimicrobial agents in animal production. One such compound may be antimicrobial peptides (AMPs), which are characterized by, among others, a wide range of biocidal activity. According to scientific data, insects produce the largest number of antimicrobial peptides, and the changing EU legislation has allowed processed animal protein derived from insects to be used in feed for farm animals, which, in addition to a protein supplement, may prove to be an alternative to antibiotics and antibiotic growth promoters due to their documented beneficial impact on livestock health. In animals that were fed feeds with the addition of insect meals, changes in their intestinal microbiota, strengthened immunity, and increased antibacterial activity were confirmed to be positive effects obtained thanks to the insect diet. This paper reviews the literature on sources of antibacterial peptides and the mechanism of action of these compounds, with particular emphasis on insect antibacterial peptides and their potential impact on animal health, and legal regulations related to the use of insect meals in animal nutrition.
Collapse
Affiliation(s)
- Ewelina Patyra
- Department of Hygiene of Animal Feedingstuffs, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland
| | - Krzysztof Kwiatek
- Department of Hygiene of Animal Feedingstuffs, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland
| |
Collapse
|
4
|
Ni M, Li W, Yuan B, Zou S, Cheng W, Yang K, Su J, Sun B, Su X. Micro-structured P-N junction surfaces: large-scale preparation, antifouling properties, and a synergistic antibacterial mechanism. J Mater Chem B 2023; 11:1312-1319. [PMID: 36651868 DOI: 10.1039/d2tb02258c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Constructing an antifouling surface cost-effectively is vitally important for many applications. Herein, a series of silicon substrates with micro-pyramid structures and p-n junctions were fabricated following a simple industrial processing flow, among which the p+n-Si substrate, fabricated through boron doping of a micro-pyramid structured n-type silicon wafer, exhibited the most pronounced antibacterial performance. Broad-spectrum bactericidal and bacteriostatic activity of p+n-Si under ambient light illumination was observed, with an inhibition ability of 73-100% compared to that of a bare glass against both airborne and contact-transmitted bacteria in the intensive care unit. The synergetic effect of mechanical rupture and electric injury was supposed to be responsible for the potent antibacterial activity. This work proposes a state-of-the-art concept that p-n junctions enhance the anti-infection ability of micro-structured surfaces and provide a promising strategy for fabricating practical antifouling surfaces with a large-size, a facile manufacturing procedure, and gentle working conditions, as well as broad-spectrum and physical antibacterial mechanisms.
Collapse
Affiliation(s)
- Mengfei Ni
- School of Physical Science and Technology, and Jiangsu Key Laboratory of Thin Films, Soochow University, Suzhou 215006, China.
| | - Wenwen Li
- School of Physical Science and Technology, and Jiangsu Key Laboratory of Thin Films, Soochow University, Suzhou 215006, China. .,Songshan Lake Materials Laboratory, Dongguan 523808, China.
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, China.
| | - Shuai Zou
- School of Physical Science and Technology, and Jiangsu Key Laboratory of Thin Films, Soochow University, Suzhou 215006, China.
| | - Wei Cheng
- School of Physical Science and Technology, and Jiangsu Key Laboratory of Thin Films, Soochow University, Suzhou 215006, China.
| | - Kai Yang
- School of Physical Science and Technology, and Jiangsu Key Laboratory of Thin Films, Soochow University, Suzhou 215006, China.
| | - Jiandong Su
- Suzhou Municipal Hospital, Suzhou 215008, China.
| | - Bingwei Sun
- Suzhou Municipal Hospital, Suzhou 215008, China.
| | - Xiaodong Su
- School of Physical Science and Technology, and Jiangsu Key Laboratory of Thin Films, Soochow University, Suzhou 215006, China.
| |
Collapse
|
5
|
Fait A, Seif Y, Mikkelsen K, Poudel S, Wells JM, Palsson BO, Ingmer H. Adaptive laboratory evolution and independent component analysis disentangle complex vancomycin adaptation trajectories. Proc Natl Acad Sci U S A 2022; 119:e2118262119. [PMID: 35858453 PMCID: PMC9335240 DOI: 10.1073/pnas.2118262119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Human infections with methicillin-resistant Staphylococcus aureus (MRSA) are commonly treated with vancomycin, and strains with decreased susceptibility, designated as vancomycin-intermediate S. aureus (VISA), are associated with treatment failure. Here, we profiled the phenotypic, mutational, and transcriptional landscape of 10 VISA strains adapted by laboratory evolution from one common MRSA ancestor, the USA300 strain JE2. Using functional and independent component analysis, we found that: 1) despite the common genetic background and environmental conditions, the mutational landscape diverged between evolved strains and included mutations previously associated with vancomycin resistance (in vraT, graS, vraFG, walKR, and rpoBCD) as well as novel adaptive mutations (SAUSA300_RS04225, ssaA, pitAR, and sagB); 2) the first wave of mutations affected transcriptional regulators and the second affected genes involved in membrane biosynthesis; 3) expression profiles were predominantly strain-specific except for sceD and lukG, which were the only two genes significantly differentially expressed in all clones; 4) three independent virulence systems (φSa3, SaeR, and T7SS) featured as the most transcriptionally perturbed gene sets across clones; 5) there was a striking variation in oxacillin susceptibility across the evolved lineages (from a 10-fold increase to a 63-fold decrease) that also arose in clinical MRSA isolates exposed to vancomycin and correlated with susceptibility to teichoic acid inhibitors; and 6) constitutive expression of the VraR regulon explained cross-susceptibility, while mutations in walK were associated with cross-resistance. Our results show that adaptation to vancomycin involves a surprising breadth of mutational and transcriptional pathways that affect antibiotic susceptibility and possibly the clinical outcome of infections.
Collapse
Affiliation(s)
- Anaëlle Fait
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, 1870 Denmark
| | - Yara Seif
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
- Merck & Co., Inc., South San Francisco, CA 94080
| | - Kasper Mikkelsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, 1870 Denmark
| | - Saugat Poudel
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Jerry M. Wells
- Host-Microbe Interactomics, Animal Sciences Group, Wageningen University, Wageningen, The Netherlands
| | - Bernhard O. Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, 1870 Denmark
| |
Collapse
|
6
|
Bucuresteanu R, Ionita M, Chihaia V, Ficai A, Trusca RD, Ilie CI, Kuncser A, Holban AM, Mihaescu G, Petcu G, Nicolaev A, Costescu RM, Husch M, Parvulescu V, Ditu LM. Antimicrobial Properties of TiO2 Microparticles Coated with Ca- and Cu-Based Composite Layers. Int J Mol Sci 2022; 23:ijms23136888. [PMID: 35805899 PMCID: PMC9266884 DOI: 10.3390/ijms23136888] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/16/2022] Open
Abstract
The ability of TiO2 to generate reactive oxygen species under UV radiation makes it an efficient candidate in antimicrobial studies. In this context, the preparation of TiO2 microparticles coated with Ca- and Cu-based composite layers over which Cu(II), Cu(I), and Cu(0) species were identified is presented here. The obtained materials were characterized by a wide range of analytical methods, such as X-ray diffraction, electron microscopy (TEM, SEM), X-ray photoelectron (XPS), and UV-VIS spectroscopy. The antimicrobial efficiency was evaluated using qualitative and quantitative standard methods and standard clinical microbial strains. A significant aspect of this composite is that the antimicrobial properties were evidenced both in the presence and absence of the light, as result of competition between photo and electrical effects. However, the antibacterial effect was similar in darkness and light for all samples. Because no photocatalytic properties were found in the absence of copper, the results sustain the antibacterial effect of the electric field (generated by the electrostatic potential of the composite layer) both under the dark and in light conditions. In this way, the composite layers supported on the TiO2 microparticles’ surface can offer continuous antibacterial protection and do not require the presence of a permanent light source for activation. However, the antimicrobial effect in the dark is more significant and is considered to be the result of the electric field effect generated on the composite layer.
Collapse
Affiliation(s)
- Razvan Bucuresteanu
- Microbiology Department, Faculty of Biology, University of Bucharest, Intr. Portocalelor 1-3, 060101 Bucharest, Romania; (R.B.); (A.-M.H.); (G.M.)
- Research Institute of the University of Bucharest, Sos. Panduri 90, 050663 Bucharest, Romania
| | - Monica Ionita
- Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania;
| | - Viorel Chihaia
- Institute of Physical Chemistry “Ilie Murgulescu”, Romanian Academy, Splaiul Independentei 202, 060021 Bucharest, Romania; (V.C.); (G.P.)
| | - Anton Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania; (A.F.); (C.-I.I.)
- National Centre for Micro and Nanomaterials and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Spl. Indendentei 313, 060042 Bucharest, Romania;
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Roxana-Doina Trusca
- National Centre for Micro and Nanomaterials and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Spl. Indendentei 313, 060042 Bucharest, Romania;
| | - Cornelia-Ioana Ilie
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gh Polizu Street, 011061 Bucharest, Romania; (A.F.); (C.-I.I.)
- National Centre for Micro and Nanomaterials and National Centre for Food Safety, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Spl. Indendentei 313, 060042 Bucharest, Romania;
| | - Andrei Kuncser
- National Institute of Materials Physics, 405A Atomistilor Street, 077125 Magurele, Romania; (A.K.); (A.N.); (R.M.C.)
| | - Alina-Maria Holban
- Microbiology Department, Faculty of Biology, University of Bucharest, Intr. Portocalelor 1-3, 060101 Bucharest, Romania; (R.B.); (A.-M.H.); (G.M.)
- Research Institute of the University of Bucharest, Sos. Panduri 90, 050663 Bucharest, Romania
| | - Grigore Mihaescu
- Microbiology Department, Faculty of Biology, University of Bucharest, Intr. Portocalelor 1-3, 060101 Bucharest, Romania; (R.B.); (A.-M.H.); (G.M.)
- Research Institute of the University of Bucharest, Sos. Panduri 90, 050663 Bucharest, Romania
| | - Gabriela Petcu
- Institute of Physical Chemistry “Ilie Murgulescu”, Romanian Academy, Splaiul Independentei 202, 060021 Bucharest, Romania; (V.C.); (G.P.)
| | - Adela Nicolaev
- National Institute of Materials Physics, 405A Atomistilor Street, 077125 Magurele, Romania; (A.K.); (A.N.); (R.M.C.)
| | - Ruxandra M. Costescu
- National Institute of Materials Physics, 405A Atomistilor Street, 077125 Magurele, Romania; (A.K.); (A.N.); (R.M.C.)
| | - Mihai Husch
- Faculty of Building Services Engineering, Technical University of Civil Engineering Bucharest, 020396 Bucharest, Romania;
| | - Viorica Parvulescu
- Institute of Physical Chemistry “Ilie Murgulescu”, Romanian Academy, Splaiul Independentei 202, 060021 Bucharest, Romania; (V.C.); (G.P.)
- Correspondence: (V.P.); (L.-M.D.); Tel.: +40-744-42-15-51 (V.P.); +40-745-67-38-22 (L.M.-D.)
| | - Lia-Mara Ditu
- Microbiology Department, Faculty of Biology, University of Bucharest, Intr. Portocalelor 1-3, 060101 Bucharest, Romania; (R.B.); (A.-M.H.); (G.M.)
- Research Institute of the University of Bucharest, Sos. Panduri 90, 050663 Bucharest, Romania
- Correspondence: (V.P.); (L.-M.D.); Tel.: +40-744-42-15-51 (V.P.); +40-745-67-38-22 (L.M.-D.)
| |
Collapse
|
7
|
Ma PY, Chong CW, Than LTL, Sulong AB, Ho KL, Neela VK, Sekawi Z, Liew YK. Impact of IsaA Gene Disruption: Decreasing Staphylococcal Biofilm and Alteration of Transcriptomic and Proteomic Profiles. Microorganisms 2022; 10:microorganisms10061119. [PMID: 35744637 PMCID: PMC9229027 DOI: 10.3390/microorganisms10061119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
Staphylococcus aureus expresses diverse proteins at different stages of growth. The immunodominant staphylococcal antigen A (IsaA) is one of the proteins that is constitutively produced by S. aureus during colonisation and infection. SACOL2584 (or isaA) is the gene that encodes this protein. It has been suggested that IsaA can hydrolyse cell walls, and there is still need to study isaA gene disruption to analyse its impact on staphylococcal phenotypes and on alteration to its transcription and protein profiles. In the present study, the growth curve in RPMI medium (which mimics human plasma), autolytic activity, cell wall morphology, fibronectin and fibrinogen adhesion and biofilm formation of S. aureus SH1000 (wildtype) was compared to that of S. aureus MS001 (isaA mutant). RNA sequencing and liquid chromatography–tandem mass spectrometry were carried out on samples of both S. aureus strains taken during the exponential growth phase, followed by bioinformatics analysis. Disruption of isaA had no obvious effect on the growth curve and autolysis ability or thickness of cell walls, but this study revealed significant strength of fibronectin adherence in S. aureus MS001. In particular, the isaA mutant formed less biofilm than S. aureus SH1000. In addition, proteomics and transcriptomics showed that the adhesin/biofilm-related genes and hemolysin genes, such as sasF, sarX and hlgC, were consistently downregulated with isaA gene disruption. The majority of the upregulated genes or proteins in S. aureus MS001 were pur genes. Taken together, this study provides insight into how isaA disruption changes the expression of other genes and has implications regarding biofilm formation and biological processes.
Collapse
Affiliation(s)
- Pei Yee Ma
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia; (L.T.L.T.); (V.K.N.); (Z.S.)
| | - Anita Binti Sulong
- Department of Medical Microbiology and Immunology, Pusat Perubatan UKM, Kuala Lumpur 56000, Malaysia;
| | - Ket Li Ho
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Vasantha Kumari Neela
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia; (L.T.L.T.); (V.K.N.); (Z.S.)
| | - Zamberi Sekawi
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia; (L.T.L.T.); (V.K.N.); (Z.S.)
| | - Yun Khoon Liew
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia;
- Correspondence:
| |
Collapse
|
8
|
Cho J, Costa SK, Wierzbicki RM, Rigby WFC, Cheung AL. The extracellular loop of the membrane permease VraG interacts with GraS to sense cationic antimicrobial peptides in Staphylococcus aureus. PLoS Pathog 2021; 17:e1009338. [PMID: 33647048 PMCID: PMC7951975 DOI: 10.1371/journal.ppat.1009338] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/11/2021] [Accepted: 01/27/2021] [Indexed: 01/27/2023] Open
Abstract
Host defense proteins (HDPs), aka defensins, are a key part of the innate immune system that functions by inserting into the bacterial membranes to form pores to kill invading and colonizing microorganisms. To ensure survival, microorganism such as S. aureus has developed survival strategies to sense and respond to HDPs. One key strategy in S. aureus is a two-component system (TCS) called GraRS coupled to an efflux pump that consists of a membrane permease VraG and an ATPase VraF, analogous to the BceRS-BceAB system of Bacillus subtilis but with distinct differences. While the 9 negatively charged amino acid extracellular loop of the membrane sensor GraS has been shown to be involved in sensing, the major question is how such a small loop can sense diverse HDPs. Mutation analysis in this study divulged that the vraG mutant phenocopied the graS mutant with respect to reduced activation of downstream effector mprF, reduction in surface positive charge and enhanced 2 hr. killing with LL-37 as compared with the parental MRSA strain JE2. In silico analysis revealed VraG contains a single 200-residue extracellular loop (EL) situated between the 7th and 8th transmembrane segments (out of 10). Remarkably, deletion of EL in VraG enhanced mprF expression, augmented surface positive charge and improved survival in LL-37 vs. parent JE2. As the EL of VraG is rich in lysine residues (16%), in contrast to a preponderance of negatively charged aspartic acid residues (3 out of 9) in the EL of GraS, we divulged the role of charge interaction by showing that K380 in the EL of VraG is an important residue that likely interacts with GraS to interfere with GraS-mediated signaling. Bacterial two-hybrid analysis also supported the interaction of EL of VraG with the EL of GraS. Collectively, we demonstrated an interesting facet of efflux pumps whereby the membrane permease disrupts HDP signaling by inhibiting GraS sensing that involves charged residues in the EL of VraG.
Collapse
Affiliation(s)
- Junho Cho
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Stephen K. Costa
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Rachel M. Wierzbicki
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
- Department of Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States of America
| | - William F. C. Rigby
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
- Department of Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States of America
| | - Ambrose L. Cheung
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
| |
Collapse
|
9
|
Li Z, Ma J, Ruan J, Zhuang X. Using Positively Charged Magnetic Nanoparticles to Capture Bacteria at Ultralow Concentration. NANOSCALE RESEARCH LETTERS 2019; 14:195. [PMID: 31165285 PMCID: PMC6548795 DOI: 10.1186/s11671-019-3005-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/07/2019] [Indexed: 05/11/2023]
Abstract
Detecting bacteria at low concentrations without time-consuming culture processes would allow rapid diagnoses. Since electrostatic attraction exists between negatively charged bacterial cells and positively charged magnetic nanoparticles (NP+), capture of bacteria holds great promise towards achieving this goal. Here, we present a rapid and highly efficient approach to capture Escherichia coli, which was used as a model for gram-negative bacteria. Capture of E. coli at very low concentrations of 10 and 100 CFU/mL using NP+ is rapidly and efficiently achieved within 1 h. Moreover, the capture efficiency of NP+ was over 90% by analyzing the number of bacterial colonies on the plate. Optical and transmission electron microscopy confirmed the bacterial capture abilities of electrically charged nanoparticles (NPs). In contrast, negatively charged magnetic nanoparticles (NP-) did not show affinities towards E. coli. These results showed that bacterial cells, such as E. coli, carry a negative charge. Unlike a ligand-dependent capture system, our designed NP+ has potentials to capture a broad range of bacteria via electrostatic attractions.
Collapse
Affiliation(s)
- Zhiming Li
- Institue of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
- Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen, 361003 Fujian China
| | - Jinyuan Ma
- Department of Pharmacy, Shanghai Dermatology Hospital, Shanghai, 200443 China
| | - Jun Ruan
- College of Life Sciences, Central China Normal University, Wuhan, 430079 China
| | - Xuan Zhuang
- Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen, 361003 Fujian China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350005 Fujian Province China
| |
Collapse
|
10
|
Shang F, Li L, Yu L, Ni J, Chen X, Xue T. Effects of stigmata maydis on the methicillin resistant Staphylococus aureusbiofilm formation. PeerJ 2019; 7:e6461. [PMID: 30828488 PMCID: PMC6396744 DOI: 10.7717/peerj.6461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/15/2019] [Indexed: 01/15/2023] Open
Abstract
Background Mastitis is an inflammatory reaction of the mammary gland tissue, which causes huge losses to dairy farms throughout the world. Staphylococcus aureus is the most frequent agent associated with this disease. Staphylococcus aureus isolates, which have the ability to form biofilms, usually lead to chronic mastitis in dairy cows. Moreover, methicillin resistance of the bacteria further complicates the treatment of this disease. Stigmata maydis (corn silk), a traditional Chinese medicine, possess many biological activities. Methods In this study, we performed antibacterial activity assays, biofilm formation assays and real-time reverse transcription PCR experiments to investigate the effect of stigmata maydis (corn silk) on biofilm formation and vancomycin susceptibility of methicillin-resistant Staphylococcus aureus (MRSA) strains isolated from dairy cows with mastitis. Results In this study, the aqueous extracts of stigmata maydis inhibited the biofilm formation ability of MRSA strains and increased the vancomycin susceptibility of the strains under biofilm-cultured conditions. Conclusion This study proves that the aqueous extracts of stigmata maydis inhibit the biofilm formation ability of MRSA strains and increase the vancomycin susceptibility of the MRSA strains under biofilm-cultured conditions.
Collapse
|
11
|
The pH-dependence of lipid-mediated antimicrobial peptide resistance in a model staphylococcal plasma membrane: A two-for-one mechanism of epithelial defence circumvention. Eur J Pharm Sci 2019; 128:43-53. [DOI: 10.1016/j.ejps.2018.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/21/2018] [Accepted: 11/18/2018] [Indexed: 11/18/2022]
|
12
|
Xu Z, Coriand L, Loeffler R, Geis-Gerstorfer J, Zhou Y, Scheideler L, Fleischer M, Gehring FK, Rupp F. Saliva-coated titanium biosensor detects specific bacterial adhesion and bactericide caused mass loading upon cell death. Biosens Bioelectron 2019; 129:198-207. [PMID: 30721795 DOI: 10.1016/j.bios.2019.01.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/07/2019] [Accepted: 01/13/2019] [Indexed: 12/11/2022]
Abstract
Bacteria adhering to implanted medical devices can cause invasive microbial infections, of e.g. skin, lung or blood. In dentistry, Streptococcus gordonii is an early oral colonizer initiating dental biofilm formation and also being involved in life-threatening infective endocarditis. To treat oral biofilms, antibacterial mouth rinses are commonly used. Such initial biomaterial-bacteria interactions and the influence of antibacterial treatments are poorly understood and investigated here in situ by quartz crystal microbalance with dissipation monitoring (QCM-D). A saliva-coated titanium (Ti) biosensor is applied to analyze possible specific signal patterns indicating microbial binding mechanisms and bactericide-caused changes in bacterial film rigidity or cell leakage caused by a clinically relevant antibacterial agent (ABA), i.e., a mouth rinse comprising chlorhexidine (CHX) and cetylpyridinium chloride (CPC). Apparent missing mass effects during the formation of microscopically proven dense and vital bacterial films indicate punctual, specific binding of S. gordonii to the saliva-coated biosensor, compared to unspecific adhesion to pure Ti. Coincidentally to ABA-induced killing of surface-adhered bacteria, an increase of adsorbed dissipative mass can be sensed, contrary to the prior mass-loss. This suggests the acoustic sensing of the leakage of cellular content caused by bacterial cell wall rupturing and membrane damage upon the bactericidal attack. The results have significant implications for testing bacterial adhesion mechanisms and cellular integrity during interaction with antibacterial agents.
Collapse
Affiliation(s)
- Zeqian Xu
- University Hospital Tübingen, Section Medical Materials Science & Technology, Osianderstr. 2-8, D-72076 Tübingen, Germany; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, PR China
| | - Luisa Coriand
- Fraunhofer Institute for Applied Optics and Precision Engineering, Albert-Einstein-Strasse 7, D-07745 Jena, Germany
| | - Ronny Loeffler
- Core Facility LISA(+), Eberhard Karls University Tübingen, Auf der Morgenstelle 15, D-72076 Tübingen, Germany
| | - Juergen Geis-Gerstorfer
- University Hospital Tübingen, Section Medical Materials Science & Technology, Osianderstr. 2-8, D-72076 Tübingen, Germany
| | - Yi Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, PR China
| | - Lutz Scheideler
- University Hospital Tübingen, Section Medical Materials Science & Technology, Osianderstr. 2-8, D-72076 Tübingen, Germany
| | - Monika Fleischer
- Core Facility LISA(+), Eberhard Karls University Tübingen, Auf der Morgenstelle 15, D-72076 Tübingen, Germany
| | | | - Frank Rupp
- University Hospital Tübingen, Section Medical Materials Science & Technology, Osianderstr. 2-8, D-72076 Tübingen, Germany.
| |
Collapse
|
13
|
Juan C, Torrens G, Barceló IM, Oliver A. Interplay between Peptidoglycan Biology and Virulence in Gram-Negative Pathogens. Microbiol Mol Biol Rev 2018; 82:e00033-18. [PMID: 30209071 PMCID: PMC6298613 DOI: 10.1128/mmbr.00033-18] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The clinical and epidemiological threat of the growing antimicrobial resistance in Gram-negative pathogens, particularly for β-lactams, the most frequently used and relevant antibiotics, urges research to find new therapeutic weapons to combat the infections caused by these microorganisms. An essential previous step in the development of these therapeutic solutions is to identify their potential targets in the biology of the pathogen. This is precisely what we sought to do in this review specifically regarding the barely exploited field analyzing the interplay among the biology of the peptidoglycan and related processes, such as β-lactamase regulation and virulence. Hence, here we gather, analyze, and integrate the knowledge derived from published works that provide information on the topic, starting with those dealing with the historically neglected essential role of the Gram-negative peptidoglycan in virulence, including structural, biogenesis, remodeling, and recycling aspects, in addition to proinflammatory and other interactions with the host. We also review the complex link between intrinsic β-lactamase production and peptidoglycan metabolism, as well as the biological costs potentially associated with the expression of horizontally acquired β-lactamases. Finally, we analyze the existing evidence from multiple perspectives to provide useful clues for identifying targets enabling the future development of therapeutic options attacking the peptidoglycan-virulence interconnection as a key weak point of the Gram-negative pathogens to be used, if not to kill the bacteria, to mitigate their capacity to produce severe infections.
Collapse
Affiliation(s)
- Carlos Juan
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| | - Gabriel Torrens
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| | - Isabel Maria Barceló
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| | - Antonio Oliver
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| |
Collapse
|
14
|
MgrA Negatively Regulates Biofilm Formation and Detachment by Repressing the Expression of psm Operons in Staphylococcus aureus. Appl Environ Microbiol 2018. [PMID: 29884758 DOI: 10.1128/aem01008-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Phenol-soluble modulins (PSMs) are amphipathic peptides that are produced by staphylococci and play important roles in Staphylococcus aureus biofilm formation and dissemination. Although the multiple functions of PSMs have been recognized, the regulatory mechanisms controlling the expression of psm operons remain largely unknown. In this study, we identified MgrA in a DNA pulldown assay and further demonstrated, by electrophoretic mobility shift assays and DNase I footprinting assays, that MgrA could bind specifically to the promoter regions of psm operons. We then constructed an isogenic mgrA deletion strain and compared biofilm formation and detachment in the wild-type and isogenic mgrA deletion strains. Our results indicated that biofilm formation and detachment were significantly increased in the mgrA mutant strain. Real-time quantitative reverse transcription-PCR data indicated that MgrA repressed the transcription of psm operons in cultures and biofilms, suggesting that MgrA is a negative regulator of psm expression. Furthermore, we analyzed biofilm formation by the psm mutant strains, and we found that PSMs promoted biofilm structuring and development in the mgrA mutant strain. These findings reveal that MgrA negatively regulates biofilm formation and detachment by repressing the expression of psm operons through direct binding to the psm promoter regions.IMPORTANCEStaphylococcus aureus is a human and animal pathogen that can cause biofilm-associated infections. PSMs have multiple functions in biofilm development and virulence in staphylococcal pathogenesis. This study has revealed that MgrA can negatively regulate psm expression by binding directly to the promoter regions of psm operons. Furthermore, our results show that MgrA can modulate biofilm structuring and development by repressing the production of PSMs in S. aureus Our findings provide novel insights into the regulatory mechanisms of S. aureus psm gene expression, biofilm development, and pathogenesis.
Collapse
|
15
|
MgrA Negatively Regulates Biofilm Formation and Detachment by Repressing the Expression of psm Operons in Staphylococcus aureus. Appl Environ Microbiol 2018; 84:AEM.01008-18. [PMID: 29884758 PMCID: PMC6070752 DOI: 10.1128/aem.01008-18] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/03/2018] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a human and animal pathogen that can cause biofilm-associated infections. PSMs have multiple functions in biofilm development and virulence in staphylococcal pathogenesis. This study has revealed that MgrA can negatively regulate psm expression by binding directly to the promoter regions of psm operons. Furthermore, our results show that MgrA can modulate biofilm structuring and development by repressing the production of PSMs in S. aureus. Our findings provide novel insights into the regulatory mechanisms of S. aureus psm gene expression, biofilm development, and pathogenesis. Phenol-soluble modulins (PSMs) are amphipathic peptides that are produced by staphylococci and play important roles in Staphylococcus aureus biofilm formation and dissemination. Although the multiple functions of PSMs have been recognized, the regulatory mechanisms controlling the expression of psm operons remain largely unknown. In this study, we identified MgrA in a DNA pulldown assay and further demonstrated, by electrophoretic mobility shift assays and DNase I footprinting assays, that MgrA could bind specifically to the promoter regions of psm operons. We then constructed an isogenic mgrA deletion strain and compared biofilm formation and detachment in the wild-type and isogenic mgrA deletion strains. Our results indicated that biofilm formation and detachment were significantly increased in the mgrA mutant strain. Real-time quantitative reverse transcription-PCR data indicated that MgrA repressed the transcription of psm operons in cultures and biofilms, suggesting that MgrA is a negative regulator of psm expression. Furthermore, we analyzed biofilm formation by the psm mutant strains, and we found that PSMs promoted biofilm structuring and development in the mgrA mutant strain. These findings reveal that MgrA negatively regulates biofilm formation and detachment by repressing the expression of psm operons through direct binding to the psm promoter regions. IMPORTANCEStaphylococcus aureus is a human and animal pathogen that can cause biofilm-associated infections. PSMs have multiple functions in biofilm development and virulence in staphylococcal pathogenesis. This study has revealed that MgrA can negatively regulate psm expression by binding directly to the promoter regions of psm operons. Furthermore, our results show that MgrA can modulate biofilm structuring and development by repressing the production of PSMs in S. aureus. Our findings provide novel insights into the regulatory mechanisms of S. aureus psm gene expression, biofilm development, and pathogenesis.
Collapse
|
16
|
Monroy GL, Hong W, Khampang P, Porter RG, Novak MA, Spillman DR, Barkalifa R, Chaney EJ, Kerschner JE, Boppart SA. Direct Analysis of Pathogenic Structures Affixed to the Tympanic Membrane during Chronic Otitis Media. Otolaryngol Head Neck Surg 2018; 159:117-126. [PMID: 29587128 DOI: 10.1177/0194599818766320] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Objective To characterize otitis media-associated structures affixed to the mucosal surface of the tympanic membrane (TM) in vivo and in surgically recovered in vitro samples. Study Design Prospective case series without comparison. Setting Outpatient surgical care center. Subjects and Methods Forty pediatric subjects scheduled for tympanostomy tube placement surgery were imaged intraoperatively under general anesthesia. Postmyringotomy, a portable optical coherence tomography (OCT) imaging system assessed for the presence of any biofilm affixed to the mucosal surface of the TM. Samples of suspected microbial infection-related structures were collected through the myringotomy incision. The sampled site was subsequently reimaged with OCT to confirm collection from the original image site on the TM. In vitro analysis based on confocal laser scanning microscope (CLSM) images of fluorescence in situ hybridization-tagged samples and polymerase chain reaction (PCR) provided microbiological characterization and verification of biofilm activity. Results OCT imaging was achieved for 38 of 40 subjects (95%). Images from 38 of 38 (100%) of subjects observed with OCT showed the presence of additional microbial infection-related structures. Thirty-four samples were collected from these 38 subjects. CLSM images provided evidence of clustered bacteria in 32 of 33 (97%) of samples. PCR detected the presence of active bacterial DNA signatures in 20 of 31 (65%) of samples. Conclusion PCR and CLSM analysis of fluorescence in situ hybridization-stained samples validates the presence of active bacteria that have formed into a middle ear biofilm that extends across the mucosal layer of the TM. OCT can rapidly and noninvasively identify middle ear biofilms in subjects with severe and persistent cases of otitis media.
Collapse
Affiliation(s)
- Guillermo L Monroy
- 1 Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,2 Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Wenzhou Hong
- 3 Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Ryan G Porter
- 4 Department of Otolaryngology-Head and Neck Surgery, Carle Foundation Hospital, Urbana, Illinois, USA.,5 Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Michael A Novak
- 4 Department of Otolaryngology-Head and Neck Surgery, Carle Foundation Hospital, Urbana, Illinois, USA.,5 Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Darold R Spillman
- 2 Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Ronit Barkalifa
- 2 Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Eric J Chaney
- 2 Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | | | - Stephen A Boppart
- 1 Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,2 Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA.,5 Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
17
|
Montanaro L, Campoccia D, Arciola CR. A Glance at the Role of Exotoxins in Opportunistic Bacterial Infections. Int J Artif Organs 2018; 29:462-7. [PMID: 16705616 DOI: 10.1177/039139880602900417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The production and the mechanism of action of exotoxins from Staphylococcus aureus, Staphylococcus epidermidis and Pseudomonas aeruginosa are presented. The attack to the immune host's defenses is the main virulence factor of opportunistic bacteria in implant infections, favoring the invasion and colonization of compromised periprosthesis tissues.
Collapse
Affiliation(s)
- L Montanaro
- Research Laboratory on Biocompatibility of Implant Materials, Rizzoli Orthopedic Institute, Bologna and Experimental Pathology Department, University of Bologna, Italy.
| | | | | |
Collapse
|
18
|
Fleming G, Aveyard J, Fothergill JL, McBride F, Raval R, D'Sa RA. Nitric Oxide Releasing Polymeric Coatings for the Prevention of Biofilm Formation. Polymers (Basel) 2017; 9:E601. [PMID: 30965904 PMCID: PMC6418929 DOI: 10.3390/polym9110601] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/30/2017] [Accepted: 11/08/2017] [Indexed: 01/14/2023] Open
Abstract
The ability of nitric oxide (NO)-releasing polymer coatings to prevent biofilm formation is described. NO-releasing coatings on (poly(ethylene terephthalate) (PET) and silicone elastomer (SE)) were fabricated using aminosilane precursors. Pristine PET and SE were oxygen plasma treated, followed by immobilisation of two aminosilane molecules: N-(3-(trimethoxysilyl)propyl)diethylenetriamine (DET3) and N-(3-trimethoxysilyl)propyl)aniline (PTMSPA). N-diazeniumdiolate nitric oxide donors were formed at the secondary amine sites on the aminosilane molecules producing NO-releasing polymeric coatings. The NO payload and release were controlled by the aminosilane precursor, as DET3 has two secondary amine sites and PTMSPA only one. The antibacterial efficacy of these coatings was tested using a clinical isolate of Pseudomonas aeruginosa (PA14). All NO-releasing coatings in this study were shown to significantly reduce P. aeruginosa adhesion over 24 h with the efficacy being a function of the aminosilane modification and the underlying substrate. These NO-releasing polymers demonstrate the potential and utility of this facile coating technique for preventing biofilms for indwelling medical devices.
Collapse
Affiliation(s)
- George Fleming
- Department of Mechanical, Materials and Aerospace Engineering, University of Liverpool, Liverpool L69 3GH, UK.
| | - Jenny Aveyard
- Department of Mechanical, Materials and Aerospace Engineering, University of Liverpool, Liverpool L69 3GH, UK.
| | - Joanne L Fothergill
- Institute of Infection and Global Health, University of Liverpool, 8 West Derby Street, Liverpool L69 7B3, UK.
| | - Fiona McBride
- The Open Innovation Hub for Antimicrobial Surfaces, Surface Science Research Centre, Department of Chemistry, University of Liverpool, Liverpool L69 3BX, UK.
| | - Rasmita Raval
- The Open Innovation Hub for Antimicrobial Surfaces, Surface Science Research Centre, Department of Chemistry, University of Liverpool, Liverpool L69 3BX, UK.
| | - Raechelle A D'Sa
- Department of Mechanical, Materials and Aerospace Engineering, University of Liverpool, Liverpool L69 3GH, UK.
| |
Collapse
|
19
|
Clonal dissemination of vancomycin-resistant Enterococcus faecium ST412 in a Brazilian region. Braz J Infect Dis 2017; 21:656-659. [PMID: 28759747 PMCID: PMC9425459 DOI: 10.1016/j.bjid.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/23/2017] [Accepted: 07/09/2017] [Indexed: 11/23/2022] Open
Abstract
Vancomycin-resistant Enterococcus faecium (VREfm) has emerged as an important global nosocomial pathogen, and this trend is associated with the spread of high-risk clones. Here, we determined the genetic and phenotypic features of 93 VREfm isolates that were obtained from patients in 13 hospitals in Vitória, Espírito Santo, Brazil, during 2012–2013. All the isolates were vancomycin-resistant and harbored the vanA gene. Only 6 (6.5%) of the VREfm isolates showed the ability to form biofilm. The 93 isolates analyzed belong to a single pulsed-field gel electrophoresis lineage and presented six subtypes. MLST genotyping showed that all VREfm belonged to ST412 (the high-risk clone, hospital-adapted). The present study describes the dissemination of ST412 clone in the local hospitals. The clonal spread of these ST412 isolates in the area we analyzed as well as other hospitals in southeastern Brazil supports the importance of identifying and controlling the presence of these microorganisms in health care-related services.
Collapse
|
20
|
Kaur G, Balamurugan P, Vasudevan S, Jadav S, Princy SA. Antimicrobial and Antibiofilm Potential of Acyclic Amines and Diamines against Multi-Drug Resistant Staphylococcus aureus. Front Microbiol 2017; 8:1767. [PMID: 28966610 PMCID: PMC5605668 DOI: 10.3389/fmicb.2017.01767] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/31/2017] [Indexed: 11/13/2022] Open
Abstract
Multi-drug resistant Staphylococcus aureus (MDRSA) remains a great challenge despite a decade of research on antimicrobial compounds against their infections. In the present study, various acyclic amines and diamines were chemically synthesized and tested for their antimicrobial as well as antibiofilm activity against MDRSA. Among all the synthesized compounds, an acyclic diamine, (2,2'-((butane-1,4-diylbis(azanediyl)bis(methylene))diphenol) designated as ADM 3, showed better antimicrobial activity (minimum inhibitory concentration at 50 μg/mL) and antibiofilm activity (MBIC50 at 5 μg/mL). In addition, ADM 3 was capable of reducing the virulence factors expression (anti-virulence). Confocal laser scanning microscope analysis of the in vitro tested urinary catheters showed biofilm reduction as well as bacterial killing by ADM 3. On the whole, our data suggest that acyclic diamines, especially ADM 3 can be a potent lead for the further studies in alternative therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - S. A. Princy
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA UniversityThanjavur, India
| |
Collapse
|
21
|
Han SM, Sorabella RA, Vasan S, Grbic M, Lambert D, Prasad R, Wang C, Kurlansky P, Borger MA, Gordon R, George I. Influence of Staphylococcus aureus on Outcomes after Valvular Surgery for Infective Endocarditis. J Cardiothorac Surg 2017; 12:57. [PMID: 28728556 PMCID: PMC5520392 DOI: 10.1186/s13019-017-0623-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 07/13/2017] [Indexed: 11/10/2022] Open
Abstract
Background As Staphylococcus aureus (SA) remains one of the leading cause of infective endocarditis (IE), this study evaluates whether S. aureus is associated with more severe infections or worsened outcomes compared to non-S. aureus (NSA) organisms. Methods All patients undergoing valve surgery for bacterial IE between 1995 and 2013 at our institution were included in this study (n = 323). Clinical data were retrospectively collected from the chart review. Patients were stratified according to the causative organism; SA (n = 85) and NSA (n = 238). Propensity score matched pairs (n = 64) of SA versus NSA were used in the analysis. Results SA patients presented with more severe IE compared to NSA patients, with higher rates of preoperative vascular complications, preoperative septic shock, preoperative embolic events, preoperative stroke, and annular abscess. Among the matched pairs, there were no significant differences in 30-day (9.4% SA vs. 7.8% NSA, OR = 1.20, p = 0.76) or 1-year mortality (20.3% SA vs. 14.1% NSA, OR = 1.57, p = 0.35) groups, though late survival was significantly worse in SA patients. There was also no significant difference in postoperative morbidity between the two matched groups. Conclusions SA IE is associated with a more severe clinical presentation than IE caused by other organisms. Despite the clearly increased preoperative risk, valvular surgery may benefit SA IE patients by moderating the post-operative mortality and morbidity.
Collapse
Affiliation(s)
- Sang Myung Han
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Robert A Sorabella
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Sowmya Vasan
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Mark Grbic
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Daniel Lambert
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Rahul Prasad
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Catherine Wang
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Paul Kurlansky
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Michael A Borger
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA
| | - Rachel Gordon
- Division of Infectious Diseases, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Isaac George
- Division of Cardiothoracic Surgery, New York Presbyterian Hospital - College of Physicians and Surgeons of Columbia University, 177 Fort Washington Ave, MHB 7GN-435, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Rasigade JP. Catching the evader: Can monoclonal antibodies interfere with Staphylococcus aureus immune escape? Virulence 2017; 9:1-4. [PMID: 28441093 PMCID: PMC5955477 DOI: 10.1080/21505594.2017.1320012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
23
|
Li Z, Hwang S, Bar-Peled M. Discovery of a Unique Extracellular Polysaccharide in Members of the Pathogenic Bacillus That Can Co-form with Spores. J Biol Chem 2016; 291:19051-67. [PMID: 27402849 DOI: 10.1074/jbc.m116.724708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Indexed: 11/06/2022] Open
Abstract
An exopolysaccharide, produced during the late stage of stationary growth phase, was discovered and purified from the culture medium of Bacillus cereus, Bacillus anthracis, and Bacillus thuringiensis when strains were grown in a defined nutrient medium that induces biofilm. Two-dimensional NMR structural characterization of the polysaccharide, named pzX, revealed that it is composed of an unusual three amino-sugar sequence repeat of [-3)XylNAc4OAc(α1-3)GlcNAcA4OAc(α1-3)XylNAc(α1-]n The sugar residue XylNAc had never been described previously in any glycan structure. The XNAC operon that contains the genes for the assembly of pzX is also unique and so far has been identified only in members of the Bacillus cereus sensu lato group. Microscopic and biochemical analyses indicate that pzX co-forms during sporulation, so that upon the release of the spore to the extracellular milieu it becomes surrounded by pzX. The relative amounts of pzX produced can be manipulated by specific nutrients in the medium, but rich medium appears to suppress pzX formation. pzX has the following unique characteristics: a surfactant property that lowers surface tension, a cell/spore antiaggregant, and an adherence property that increases spores binding to surfaces. pzX in Bacillus could represent a trait shared by many spore-producing microorganisms. It suggests pzX is an active player in spore physiology and may provide new insights to the successful survival of the B. cereus species in natural environments or in the hosts.
Collapse
Affiliation(s)
- Zi Li
- From the Complex Carbohydrate Research Center and Department of Plant Biology, University of Georgia, Athens, Georgia 30602
| | - Soyoun Hwang
- From the Complex Carbohydrate Research Center and
| | - Maor Bar-Peled
- From the Complex Carbohydrate Research Center and Department of Plant Biology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
24
|
Mycobacteriophage putative GTPase-activating protein can potentiate antibiotics. Appl Microbiol Biotechnol 2016; 100:8169-77. [PMID: 27345061 DOI: 10.1007/s00253-016-7681-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/09/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
The soaring incidences of infection by antimicrobial resistant (AR) pathogens and shortage of effective antibiotics with new mechanisms of action have renewed interest in phage therapy. This scenario is exemplified by resistant tuberculosis (TB), caused by resistant Mycobacterium tuberculosis. Mycobacteriophage SWU1 A321_gp67 encodes a putative GTPase-activating protein. Mycobacterium smegmatis with gp67 overexpression showed changed colony formation and biofilm morphology and supports the efficacy of streptomycin and capreomycin against Mycobacterium. gp67 down-regulated the transcription of genes involved in cell wall and biofilm development. To our knowledge, this is the first report to show that phage protein in addition to lysin or recombination components can synergize with existing antibiotics. Phage components might represent a promising new clue for better antibiotic potentiators.
Collapse
|
25
|
Krismastuti FSH, Cavallaro A, Prieto-Simon B, Voelcker NH. Toward Multiplexing Detection of Wound Healing Biomarkers on Porous Silicon Resonant Microcavities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2016; 3:1500383. [PMID: 27812471 PMCID: PMC5067563 DOI: 10.1002/advs.201500383] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 12/17/2015] [Indexed: 06/06/2023]
Abstract
Bacterial wound infections can cause septicemia and lead to limb amputation or death. Therefore, early detection of bacteria is important in chronic wound management. Here, an optical biosensor based on porous silicon resonant microcavity (pSiRM) structure modified with fluorogenic peptide substrate is demonstrated to detect the presence of Sortase A (SrtA), a bacterial enzyme found in the cell membrane protein of Staphylococcus aureus. The combination of fluorescence enhancement effects of the pSiRM architecture with the incorporation of SrtA fluorogenic peptide substrate within the pSi matrix enables the sensing of SrtA with an outstanding limit of detection of 8 × 10-14 m. Modification of the pSiRM structure with microscale spots of two fluorogenic peptide substrates, one specific for SrtA and the other for matrix metalloproteinases, effectively demonstrates the feasibility to perform multiplexed biomarker analysis. The results in this study highlight the potential of the pSiRM sensing platform as a point-of-care diagnostic tool for biomarkers of bacterial wound infection.
Collapse
Affiliation(s)
- Fransiska Sri Herwahyu Krismastuti
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology Future Industries Institute University of South Australia Mawson Lakes, Adelaide South Australia 5095 Australia
| | - Alex Cavallaro
- Future Industries Institute University of South Australia Mawson Lakes South Australia 5095 Australia
| | - Beatriz Prieto-Simon
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology Future Industries Institute University of South Australia Mawson Lakes, Adelaide South Australia 5095 Australia
| | - Nicolas H Voelcker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology Future Industries Institute University of South Australia Mawson Lakes, Adelaide South Australia 5095 Australia
| |
Collapse
|
26
|
Ost M, Singh A, Peschel A, Mehling R, Rieber N, Hartl D. Myeloid-Derived Suppressor Cells in Bacterial Infections. Front Cell Infect Microbiol 2016; 6:37. [PMID: 27066459 PMCID: PMC4814452 DOI: 10.3389/fcimb.2016.00037] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/15/2016] [Indexed: 01/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) comprise monocytic and granulocytic innate immune cells with the capability of suppressing T- and NK-cell responses. While the role of MDSCs has been studied in depth in malignant diseases, the understanding of their regulation and function in infectious disease conditions has just begun to evolve. Here we summarize and discuss the current view how MDSCs participate in bacterial infections and how this knowledge could be exploited for potential future therapeutics.
Collapse
Affiliation(s)
- Michael Ost
- Children's Hospital, University of Tübingen Tübingen, Germany
| | - Anurag Singh
- Children's Hospital, University of Tübingen Tübingen, Germany
| | - Andreas Peschel
- Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| | - Roman Mehling
- Children's Hospital, University of Tübingen Tübingen, Germany
| | - Nikolaus Rieber
- Children's Hospital, University of TübingenTübingen, Germany; Department of Pediatrics, Kinderklinik München Schwabing, Klinikum Schwabing, StKM GmbH und Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany
| | - Dominik Hartl
- Children's Hospital, University of Tübingen Tübingen, Germany
| |
Collapse
|
27
|
Edwards K. New Twist on an Old Favorite: Gentian Violet and Methylene Blue Antibacterial Foams. Adv Wound Care (New Rochelle) 2016; 5:11-18. [PMID: 26858911 DOI: 10.1089/wound.2014.0593] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Significance: Absorptive antibacterial dressings that assist in controlling bioburden without risks of cytotoxicity or residual absorption can be effectively used for prolonged periods throughout the wound healing continuum. Recent Advances: Until recently, gentian violet and methylene blue (GV/MB) antibacterial dressings have been commercially available only in polyvinyl alcohol (PVA) foam; polyurethane (PU) foam bonded with GV and MB with thin film backing is now commercially available. GV/MB PU foam does not require hydration or a necessary secondary dressing. GV/MB PVA and PU foam dressings were recently granted FDA clearance as antibacterial dressings, as opposed to bacteriostatic dressings as previously classified. Within the class of antibacterial dressings, GV/MB foam dressings are of lower cost alternative to silver- or iodine-based antibacterial dressings with no risk of absorption of any of the foam components into the tissues. Critical Issues: Control of wound bioburden levels by antibacterial agents and absorption of excess exudate are crucial in preventing infections that drastically increase the price of wound care. Use of GV/MB dressings may improve wound healing outcomes and decrease overall costs through super absorption, promotion of autolytic debridement, bioburden reduction, ease of use, and decreased dressing change frequency. Future Directions: Evolution in resistant bacterial strains will drive continual changes in advanced wound care products. Demand will increase for economically priced, versatile wound care dressings that assist in debridement, maintain a moist wound environment, absorb and trap bacterial debris, and decrease dressing change frequency.
Collapse
Affiliation(s)
- Karen Edwards
- University of Alabama Hospital at Birmingham, Birmingham, Alabama
| |
Collapse
|
28
|
Kaneti G, Meir O, Mor A. Controlling bacterial infections by inhibiting proton-dependent processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:995-1003. [PMID: 26522076 DOI: 10.1016/j.bbamem.2015.10.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022]
Abstract
Bacterial resistance to antibiotics is recognized as one of the greatest threats in modern healthcare, taking a staggering toll worldwide. New approaches for controlling bacterial infections must be designed, eventually combining multiple strategies for complimentary therapies. This review explores an old/new paradigm for multi-targeted antibacterial therapy, focused at disturbing bacterial cytoplasmic membrane functions at sub minimal inhibitory concentrations, namely through superficial physical alterations of the bilayer, thereby perturbing transmembrane signals transduction. Such a paradigm may have the advantage of fighting the infection while avoiding many of the known resistance mechanisms. This article is part of a Special Issue entitled: Antimicrobial peptides edited by Karl Lohner and Kai Hilpert.
Collapse
Affiliation(s)
- Galoz Kaneti
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ohad Meir
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Amram Mor
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
29
|
Chen X, Shang F, Meng Y, Li L, Cui Y, Zhang M, Qi K, Xue T. Ethanol extract of Sanguisorba officinalis L. inhibits biofilm formation of methicillin-resistant Staphylococcus aureus in an ica-dependent manner. J Dairy Sci 2015; 98:8486-91. [PMID: 26454299 DOI: 10.3168/jds.2015-9899] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/22/2015] [Indexed: 11/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an important nosocomial pathogen that shows resistance to many antibiotics and is usually associated with serious infections. Having the ability for biofilm formation increases resistance to antibiotics. Sanguisorba officinalis L. is a perennial plant that is distributed in the northern districts of China and has been used as a traditional Chinese medicine. In this study, the effect of S. officinalis on MRSA strain SA3 isolated from a dairy cow with mastitis was evaluated by testing the growth and biofilm formation ability of MRSA cultured with or without ethanol extracts of S. officinalis. The results showed that the ethanol extract of S. officinalis strongly inhibited the biofilm formation of MRSA. With a confocal laser scanning microscope system, we observed that the biofilm structure of the test group with the addition of S. officinalis appeared looser and had less biomass compared with the control group without S. officinalis. Furthermore, we found that the transcript levels of the icaADBC operon remarkably decreased upon addition of the ethanol extract of S. officinalis, indicating that S. officinalis inhibits biofilm formation of MRSA in an ica-dependent manner.
Collapse
Affiliation(s)
- Xiaolin Chen
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Fei Shang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yajing Meng
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Long Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunmei Cui
- School of Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ming Zhang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Kezong Qi
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
30
|
Hansmann B, Schröder JM, Gerstel U. Skin-Derived C-Terminal Filaggrin-2 Fragments Are Pseudomonas aeruginosa-Directed Antimicrobials Targeting Bacterial Replication. PLoS Pathog 2015; 11:e1005159. [PMID: 26371476 PMCID: PMC4570713 DOI: 10.1371/journal.ppat.1005159] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/20/2015] [Indexed: 01/08/2023] Open
Abstract
Soil- and waterborne bacteria such as Pseudomonas aeruginosa are constantly challenging body surfaces. Since infections of healthy skin are unexpectedly rare, we hypothesized that the outermost epidermis, the stratum corneum, and sweat glands directly control the growth of P. aeruginosa by surface-provided antimicrobials. Due to its high abundance in the upper epidermis and eccrine sweat glands, filaggrin-2 (FLG2), a water-insoluble 248 kDa S100 fused-type protein, might possess these innate effector functions. Indeed, recombinant FLG2 C-terminal protein fragments display potent antimicrobial activity against P. aeruginosa and other Pseudomonads. Moreover, upon cultivation on stratum corneum, P. aeruginosa release FLG2 C-terminus-containing FLG2 fragments from insoluble material, indicating liberation of antimicrobially active FLG2 fragments by the bacteria themselves. Analyses of the underlying antimicrobial mechanism reveal that FLG2 C-terminal fragments do not induce pore formation, as known for many other antimicrobial peptides, but membrane blebbing, suggesting an alternative mode of action. The association of the FLG2 fragment with the inner membrane of treated bacteria and its DNA-binding implicated an interference with the bacterial replication that was confirmed by in vitro and in vivo replication assays. Probably through in situ-activation by soil- and waterborne bacteria such as Pseudomonads, FLG2 interferes with the bacterial replication, terminates their growth on skin surface and thus may contributes to the skin's antimicrobial defense shield. The apparent absence of FLG2 at certain body surfaces, as in the lung or of burned skin, would explain their higher susceptibility towards Pseudomonas infections and make FLG2 C-terminal fragments and their derivatives candidates for new Pseudomonas-targeting antimicrobials.
Collapse
Affiliation(s)
- Britta Hansmann
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Ulrich Gerstel
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
31
|
Affiliation(s)
- Eric Kong
- Graduate Program in Life Sciences, Molecular Microbiology and Immunology Program, University of Maryland, Baltimore, Maryland, United States of America
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, United States of America
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
32
|
Serra R, Grande R, Butrico L, Rossi A, Settimio UF, Caroleo B, Amato B, Gallelli L, de Franciscis S. Chronic wound infections: the role of Pseudomonas aeruginosa and Staphylococcus aureus. Expert Rev Anti Infect Ther 2015; 13:605-13. [PMID: 25746414 DOI: 10.1586/14787210.2015.1023291] [Citation(s) in RCA: 424] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic leg ulcers affect 1-2% of the general population and are related to increased morbidity and health costs. Staphylococcus aureus and Pseudomonas aeruginosa are the most common bacteria isolated from chronic wounds. They can express virulence factors and surface proteins affecting wound healing. The co-infection of S. aureus and P. aeruginosa is more virulent than single infection. In particular, S. aureus and P. aeruginosa have both intrinsic and acquired antibiotic resistance, making clinical management of infection a real challenge, particularly in patients with comorbidity. Therefore, a correct and prompt diagnosis of chronic wound infection requires a detailed knowledge of skin bacterial flora. This is a necessary prerequisite for tailored pharmacological treatment, improving symptoms, and reducing side effects and antibiotic resistance.
Collapse
Affiliation(s)
- Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Headquarters: University Magna Græcia of Catanzaro, Viale Europa 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Monroy GL, Shelton RL, Nolan RM, Nguyen CT, Novak MA, Hill MC, McCormick DT, Boppart SA. Noninvasive depth-resolved optical measurements of the tympanic membrane and middle ear for differentiating otitis media. Laryngoscope 2015; 125:E276-82. [PMID: 25599652 DOI: 10.1002/lary.25141] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/17/2014] [Accepted: 12/15/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE/HYPOTHESIS In this study, optical coherence tomography (OCT) is used to noninvasively and quantitatively determine tympanic membrane (TM) thickness and the presence and thickness of any middle-ear biofilm located behind the TM. These new metrics offer the potential to differentiate normal, acute, and chronic otitis media (OM) infections in pediatric subjects. STUDY DESIGN Case series with comparison group. METHODS The TM thickness of 34 pediatric subjects was acquired using a custom-built, handheld OCT system following a traditional otoscopic ear exam. RESULTS Overall thickness (TM and any associated biofilm) was shown to be statistically different for normal, acute, and chronic infection groups (normal-acute and normal-chronic: P value < 0.001; acute-chronic: P value = 0.0016). Almost all observed scans from the chronic group had an accompanying biofilm structure. When the thickness of the TM and biofilm were considered separately in chronic OM, the chronic TM thickness correlated with the normal group (P value = 0.68) yet was still distinct from the acute OM group (P value < 0.001), indicating that the TM in chronic OM returns to relatively normal thickness levels. CONCLUSION Identifying these physical changes in vivo provides new metrics for noninvasively and quantitatively differentiating normal, acute, and chronic OM. This new diagnostic information has the potential to assist physicians to more effectively and efficiently screen, manage, and refer patients based on quantitative data. LEVEL OF EVIDENCE 4.
Collapse
Affiliation(s)
- Guillermo L Monroy
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A
| | - Ryan L Shelton
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A
| | - Ryan M Nolan
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A
| | - Cac T Nguyen
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A
| | - Michael A Novak
- Department of Surgery, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Department of Otolaryngology, Urbana, Illinois, U.S.A
| | - Malcolm C Hill
- Department of Pediatrics, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Department of Pediatrics, Urbana, Illinois, U.S.A
| | - Daniel T McCormick
- Carle Foundation Hospital, Urbana, Illinois, U.S.A.,Advanced MEMS, San Francisco, California, U.S.A
| | - Stephen A Boppart
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.,Department of Internal Medicine, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A
| |
Collapse
|
34
|
Tebartz C, Horst SA, Sparwasser T, Huehn J, Beineke A, Peters G, Medina E. A major role for myeloid-derived suppressor cells and a minor role for regulatory T cells in immunosuppression during Staphylococcus aureus infection. THE JOURNAL OF IMMUNOLOGY 2014; 194:1100-11. [PMID: 25548227 DOI: 10.4049/jimmunol.1400196] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Staphylococcus aureus can cause difficult-to-treat chronic infections. We recently reported that S. aureus chronic infection was associated with a profound inhibition of T cell responses. In this study, we investigated the mechanisms responsible for the suppression of T cell responses during chronic S. aureus infection. Using in vitro coculture systems, as well as in vivo adoptive transfer of CFSE-labeled OT-II cells, we demonstrated the presence of immunosuppressive mechanisms in splenocytes of S. aureus-infected mice that inhibited the response of OT-II cells to cognate antigenic stimulation. Immunosuppression was IL-10/TGF-β independent but required cell-cell proximity. Using DEREG and Foxp3(gfp) mice, we demonstrated that CD4(+)CD25(+)Foxp3(+) regulatory T cells contributed, but only to a minor degree, to bystander immunosuppression. Neither regulatory B cells nor tolerogenic dendritic cells contributed to immunosuppression. Instead, we found a significant expansion of granulocytic (CD11b(+)Ly6G(+)Ly6C(low)) and monocytic (CD11b(+)Ly6G(-)Ly6C(high)) myeloid-derived suppressor cells (MDSC) in chronically infected mice, which exerted a strong immunosuppressive effect on T cell responses. Splenocytes of S. aureus-infected mice lost most of their suppressive activity after the in vivo depletion of MDSC by treatment with gemcitabine. Furthermore, a robust negative correlation was observed between the degree of T cell inhibition and the number of MDSC. An increase in the numbers of MDSC in S. aureus-infected mice by adoptive transfer caused a significant exacerbation of infection. In summary, our results indicate that expansion of MDSC and, to a minor degree, of regulatory T cells in S. aureus-infected mice may create an immunosuppressive environment that sustains chronic infection.
Collapse
Affiliation(s)
- Christina Tebartz
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Sarah Anita Horst
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, Twincore, Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; and
| | - Georg Peters
- Institute of Medical Microbiology, University Hospital of 48149 Münster, Münster, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| |
Collapse
|
35
|
Miguel SP, Ribeiro MP, Brancal H, Coutinho P, Correia IJ. Thermoresponsive chitosan–agarose hydrogel for skin regeneration. Carbohydr Polym 2014; 111:366-73. [DOI: 10.1016/j.carbpol.2014.04.093] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/24/2014] [Accepted: 04/27/2014] [Indexed: 12/17/2022]
|
36
|
Otto M. Physical stress and bacterial colonization. FEMS Microbiol Rev 2014; 38:1250-70. [PMID: 25212723 DOI: 10.1111/1574-6976.12088] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 09/01/2014] [Accepted: 09/05/2014] [Indexed: 02/07/2023] Open
Abstract
Bacterial surface colonizers are subject to a variety of physical stresses. During the colonization of human epithelia such as on the skin or the intestinal mucosa, bacteria mainly have to withstand the mechanical stress of being removed by fluid flow, scraping, or epithelial turnover. To that end, they express a series of molecules to establish firm attachment to the epithelial surface, such as fibrillar protrusions (pili) and surface-anchored proteins that bind to human matrix proteins. In addition, some bacteria--in particular gut and urinary tract pathogens--use internalization by epithelial cells and other methods such as directed inhibition of epithelial turnover to ascertain continued association with the epithelial layer. Furthermore, many bacteria produce multilayered agglomerations called biofilms with a sticky extracellular matrix, providing additional protection from removal. This review will give an overview over the mechanisms human bacterial colonizers have to withstand physical stresses with a focus on bacterial adhesion.
Collapse
Affiliation(s)
- Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Li YW, Liu L, Huang PR, Fang W, Luo ZP, Peng HL, Wang YX, Li AX. Chronic streptococcosis in Nile tilapia, Oreochromis niloticus (L.), caused by Streptococcus agalactiae. JOURNAL OF FISH DISEASES 2014; 37:757-763. [PMID: 23952820 DOI: 10.1111/jfd.12146] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 05/27/2013] [Accepted: 05/28/2013] [Indexed: 06/02/2023]
Affiliation(s)
- Y W Li
- Key Laboratory for Aquatic Products Safety Department of the Ministry of Education/State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Akinkunmi EO, Adeyemi OI, Igbeneghu OA, Olaniyan EO, Omonisi AE, Lamikanra A. The pathogenicity of Staphylococcus epidermidis on the intestinal organs of rats and mice: an experimental investigation. BMC Gastroenterol 2014; 14:126. [PMID: 25016472 PMCID: PMC4105098 DOI: 10.1186/1471-230x-14-126] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 07/03/2014] [Indexed: 11/20/2022] Open
Abstract
Background Staphylococcus epidermidis is the most frequently isolated species of the coagulase negative staphylococci from human stool. However, it is not clear how its presence in the gut affects the cellular structures and functions of this organ. In this study therefore, the pathogenicity of strains of S. epidermidis which were isolated from the stool samples of apparently healthy children was investigated in mice and rats. Methods The albino mice (22—30 g) and albino rats (100-155 g) of both sexes were infected orally and intraperitoneally with graded doses of the bacteria and subjected to behavioral and histopathological examinations. Results Acute infection in these animals caused temporary behavioural changes as shown by restlessness and abdominal stretchings but did not result in death even at a dosage of 2 × 109 cfu/kg. Daily administration of the same dose for 14 days resulted in the death of 11 out of 21 (52.4%) mice. Histopathological examination of the affected organs showed congestions, aggregations and multinucleated hepatocytes in the liver, infiltration of the kidney tubule interstitial by chronic inflammatory cells, coagulative necrosis of the kidney, spleen, intestine and stomach cells as well as marked stroma fibrosis of the spleen. Coagulative necrosis of cells was the most frequently occurring pathological alteration. Lethality and pathological effects reflected the virulence factors expressed by the organism which are biofilm formation, haemagglutination properties and capsule production. Conclusions The results indicate that strains of S. epidermidis colonising the gut can cause serious pathological changes on certain organs such as kidney, liver, intestine, stomach and spleen which, depending on their severity, could be fatal.
Collapse
|
39
|
Almohamad S, Somarajan SR, Singh KV, Nallapareddy SR, Murray BE. Influence of isolate origin and presence of various genes on biofilm formation by Enterococcus faecium. FEMS Microbiol Lett 2014; 353:151-6. [PMID: 24606170 DOI: 10.1111/1574-6968.12418] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 01/08/2014] [Accepted: 03/04/2014] [Indexed: 11/28/2022] Open
Abstract
Enterococcus faecium, a major cause of nosocomial infections, is often isolated from conditions where biofilm is considered to be important in the establishment of infections. We investigated biofilm formation among E. faecium isolates from diverse sources and found that the occurrence and amount of biofilm formation were significantly greater in clinical isolates than fecal isolates from community volunteers. We also found that the presence of the empfm (E. faecium pilus) operon was associated with the amount of biofilm formation. Furthermore, we analyzed the possible association between the distribution of 16 putative virulence genes and the occurrence of biofilm production. Even though the prevalence of these virulence genes was significantly higher in clinical isolates, we did not observe any correlation with the occurrence of biofilm formation.
Collapse
Affiliation(s)
- Sam Almohamad
- Jordan University of Science and Technology, Irbid, Jordan
| | | | | | | | | |
Collapse
|
40
|
Zecconi A, Scali F. Staphylococcus aureus virulence factors in evasion from innate immune defenses in human and animal diseases. Immunol Lett 2013; 150:12-22. [PMID: 23376548 DOI: 10.1016/j.imlet.2013.01.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/09/2012] [Accepted: 01/08/2013] [Indexed: 01/04/2023]
Abstract
In the last decades, Staphylococcus aureus acquired a dramatic relevance in human and veterinary medicine for different reasons, one of them represented by the increasing prevalence of antibiotic resistant strains. However, antibiotic resistance is not the only weapon in the arsenal of S. aureus. Indeed, these bacteria have plenty of virulence factors, including a vast ability to evade host immune defenses. The innate immune system represents the first line of defense against invading pathogens. This system consists of three major effector mechanisms: antimicrobial peptides and enzymes, the complement system and phagocytes. In this review, we focused on S. aureus virulence factors involved in the immune evasion in the first phases of infection: TLR recognition avoidance, adhesins affecting immune response and resistance to host defenses peptides and polypeptides. Studies of innate immune defenses and their role against S. aureus are important in human and veterinary medicine given the problems related to S. aureus antimicrobial resistance. Moreover, due to the pathogen ability to manipulate the immune response, these data are needed to develop efficacious vaccines or molecules against S. aureus.
Collapse
Affiliation(s)
- Alfonso Zecconi
- Università degli Studi di Milano, Dip. Scienze Veterinarie e Sanità Pubblica, Via Celoria 10, 20133 Milano, Italy.
| | | |
Collapse
|
41
|
McNicholaswhj S, Humphreys H, Hughes DF. Susceptibility of <i>Staphylococcus aureus</i> Isolates Causing Bloodstream Infection to the Antimicrobial Peptide LL-37. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojmm.2013.31012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
42
|
Abstract
Bacteria living as biofilms have been recognised as the ultimate cause of persistent and destructive inflammatory processes. Biofilm formation is a well-organised, genetically-driven process, which is well characterised for numerous bacteria species. In contrast, the host response to bacterial biofilms is less well analysed, and there is the general believe that bacteria in biofilms escape recognition or eradication by the immune defence. In this review the host response to bacterial biofilms is discussed with particular focus on the role of neutrophils because these phagocytic cells are the first to infiltrate areas of bacterial infection, and because neutrophils are equipped with a wide arsenal of bactericidal and toxic entities. I come to the conclusion that bacterial biofilms are not inherently protected against the attack by neutrophils, but that control of biofilm formation is possible depending on a timely and sufficient host response.
Collapse
|
43
|
Passariello C, Lucchese A, Virga A, Pera F, Gigola P. Isolation of Staphylococcus Aureus and Progression of Periodontal Lesions in Aggressive Periodontitis. EUR J INFLAMM 2012. [DOI: 10.1177/1721727x1201000326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This work evaluates whether isolation and toxicity of subgingival Staphylococcus aureus strains correlate with progression of periodontal lesions and whether isolates are characterized by a specific genomic background. The study involved 165 subjects affected by generalized aggressive periodontitis. Three sets of samples of supragingival and subgingival plaque were obtained at 45-day intervals from active and non-active sites, to detect S. aureus. Susceptibility to antibiotics, the presence of 17 genes, genomic restriction profiles and multilocus sequence typing (MLST) were performed to characterize all isolates. S. aureus was detected in 37.6% of the subjects. Subgingival colonization rates were 66.1% and 12.9% for active and non-active sites, respectively ( P<0.01). Supragingival and subgingival isolates were shown to be distinct by molecular genotyping and DNA fingerprint analysis. MLST showed that isolates were not genetically related and no sequence type was predominant in any of the two locations. These data demonstrate that S. aureus is associated with the progression of aggressive periodontitis and that a specific set of characters is necessary for the bacterium to colonize subgingival sites. Comparative analysis of genomic structure and genetic-related data suggest that the periodontal environment could promote genetic evolution of strains.
Collapse
Affiliation(s)
- C. Passariello
- Dipartimento di Sanità Pubblica e Malattie Infettive, Università “Sapienza”, Roma, Italy
| | - A. Lucchese
- Dipartimento di Chirurgia Maxillofacciale, Università di Ferrara, Ferrara, Italy
| | - A. Virga
- Dipartimento di Sanità Pubblica e Malattie Infettive, Università “Sapienza”, Roma, Italy
| | - F. Pera
- Dipartimento di Implantologia e Protesi Dentale, Università di Torino, Torino, Italy
| | - P. Gigola
- Dipartimento di Specialità Chirurgiche, Scienze Radiologiche e Medico-Forensi, Università di Brescia, Italy
| |
Collapse
|
44
|
SUDAGIDAN MERT, YEMENİCİOĞLU AHMET. Effects of Nisin and Lysozyme on Growth Inhibition and Biofilm Formation Capacity of Staphylococcus aureus Strains Isolated from Raw Milk and Cheese Samples. J Food Prot 2012; 75:1627-33. [DOI: 10.4315/0362-028x.jfp-12-001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Effects of nisin and lysozyme on growth inhibition and biofilm formation capacity of 25 Staphylococcus aureus strains isolated from raw milk (13 strains) and cheese (12 strains) were studied. Nisin was tested at concentrations between 0.5 and 25 μg/ml; the growth of all strains was inhibited at 25 μg/ml, but the resistances of strains showed a great variation at lower nisin concentrations. In contrast, lysozyme tested at concentrations up to 5.0 mg/ml showed no inhibition on the growth of strains. Nisin used at the growth inhibitory concentration prevented the biofilm formation of strains, but strains continued biofilm formation at subinhibitory nisin concentrations. Lysozyme did not affect the biofilm formation of 19 of the strains, but it caused a considerable activation in the biofilm formation capacity of six strains. Twelve of the strains contained both biofilm-related protease genes (sspA, sspB, and aur) and active proteases; eight of these strains were nisin resistant. These results suggest a potential risk of S. aureus growth and biofilm formation when lysozyme is used in the biopreservation of dairy products. Nisin can be used to control growth and biofilm formation of foodborne S. aureus, unless resistance against this biopreservative develops.
Collapse
Affiliation(s)
- MERT SUDAGIDAN
- 1Science and Technology Application and Research Center, Mehmet Akif Ersoy University, 15100 Burdur, Turkey
| | - AHMET YEMENİCİOĞLU
- 2Department of Food Engineering, Izmir Institute of Technology, Gulbahce Campus, Urla 35430, Izmir, Turkey
| |
Collapse
|
45
|
Lebreton F, van Schaik W, Sanguinetti M, Posteraro B, Torelli R, Le Bras F, Verneuil N, Zhang X, Giard JC, Dhalluin A, Willems RJL, Leclercq R, Cattoir V. AsrR is an oxidative stress sensing regulator modulating Enterococcus faecium opportunistic traits, antimicrobial resistance, and pathogenicity. PLoS Pathog 2012; 8:e1002834. [PMID: 22876178 PMCID: PMC3410868 DOI: 10.1371/journal.ppat.1002834] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/18/2012] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress serves as an important host/environmental signal that triggers a wide range of responses in microorganisms. Here, we identified an oxidative stress sensor and response regulator in the important multidrug-resistant nosocomial pathogen Enterococcus faecium belonging to the MarR family and called AsrR (antibiotic and stress response regulator). The AsrR regulator used cysteine oxidation to sense the hydrogen peroxide which results in its dissociation to promoter DNA. Transcriptome analysis showed that the AsrR regulon was composed of 181 genes, including representing functionally diverse groups involved in pathogenesis, antibiotic and antimicrobial peptide resistance, oxidative stress, and adaptive responses. Consistent with the upregulated expression of the pbp5 gene, encoding a low-affinity penicillin-binding protein, the asrR null mutant was found to be more resistant to β-lactam antibiotics. Deletion of asrR markedly decreased the bactericidal activity of ampicillin and vancomycin, which are both commonly used to treat infections due to enterococci, and also led to over-expression of two major adhesins, acm and ecbA, which resulted in enhanced in vitro adhesion to human intestinal cells. Additional pathogenic traits were also reinforced in the asrR null mutant including greater capacity than the parental strain to form biofilm in vitro and greater persistance in Galleria mellonella colonization and mouse systemic infection models. Despite overexpression of oxidative stress-response genes, deletion of asrR was associated with a decreased oxidative stress resistance in vitro, which correlated with a reduced resistance to phagocytic killing by murine macrophages. Interestingly, both strains showed similar amounts of intracellular reactive oxygen species. Finally, we observed a mutator phenotype and enhanced DNA transfer frequencies in the asrR deleted strain. These data indicate that AsrR plays a major role in antimicrobial resistance and adaptation for survival within the host, thereby contributes importantly to the opportunistic traits of E. faecium. Multiple antibiotic-resistant isolates of the opportunistic pathogen Enterococcus faecium have emerged and spread worldwide. However, studies aimed at identifying mechanisms that underlie the transformation of E. faecium from its commensal nature into a nosocomial pathogen are scarce. We report pleiotropic roles for a novel oxidative-sensing regulator, called AsrR (antibiotic and stress response regulator), in E. faecium. Based on transcriptomic analysis, phenotypic studies, and animal models, we demonstrate that asrR deletion is responsible for i) diminished susceptibility to penicillins, vancomycin, and cationic antimicrobial peptides, ii) increased adhesion to human cells and biofilm formation, iii) a mutator phenotype and enhanced DNA transfer frequencies, iv) decreased resistance to oxidative stress both in vitro and in murine macrophages, and v) increased host-persistence in both insect and mouse models. AsrR is a stress-sensor and is promptly inactivated in the presence of hydrogen peroxide. Therefore, oxidative stress, which is a main challenge during infection, may be a significant signal used by E. faecium to promote opportunistic traits. This provides a significant resource combining, for the first time in E. faecium, a global transcriptomic approach and a thorough phenotypic study, which places AsrR as a key regulator modulating pathogenicity, antimicrobial resistance, and environmental adaptation.
Collapse
Affiliation(s)
- François Lebreton
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Willem van Schaik
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | | | | | - Riccardo Torelli
- Catholic University of Sacred Heart, Institute of Microbiology, Rome, Italy
| | - Florian Le Bras
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Nicolas Verneuil
- University of Caen Basse-Normandie, EA4655 (team “Stress and Virulence”), Caen, France
| | - Xinglin Zhang
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Jean-Christophe Giard
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Anne Dhalluin
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
| | - Rob J. L. Willems
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Roland Leclercq
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
- University Hospital of Caen, Department of Microbiology, Caen, France
| | - Vincent Cattoir
- University of Caen Basse-Normandie, EA4655 (team “Antibioresistance”), Medical School, Caen, France
- University Hospital of Caen, Department of Microbiology, Caen, France
- * E-mail:
| |
Collapse
|
46
|
WIPI-1 Positive Autophagosome-Like Vesicles Entrap Pathogenic Staphylococcus aureus for Lysosomal Degradation. Int J Cell Biol 2012; 2012:179207. [PMID: 22829830 PMCID: PMC3399381 DOI: 10.1155/2012/179207] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/31/2012] [Accepted: 02/06/2012] [Indexed: 12/27/2022] Open
Abstract
Invading pathogens provoke the autophagic machinery and, in a process termed xenophagy, the host cell survives because autophagy is employed as a safeguard for pathogens that escaped phagosomes. However, some pathogens can manipulate the autophagic pathway and replicate within the niche of generated autophagosome-like vesicles. By automated fluorescence-based high content analyses, we demonstrate that Staphylococcus aureus strains (USA300, HG001, SA113) stimulate autophagy and become entrapped in intracellular PtdIns(3)P-enriched vesicles that are decorated with human WIPI-1, an essential PtdIns(3)P effector of canonical autophagy and membrane protein of both phagophores and autophagosomes. Further, agr-positive S. aureus (USA300, HG001) strains were more efficiently entrapped in WIPI-1 positive autophagosome-like vesicles when compared to agr-negative cells (SA113). By confocal and electron microscopy we provide evidence that single- and multiple-Staphylococci entrapped undergo cell division. Moreover, the number of WIPI-1 positive autophagosome-like vesicles entrapping Staphylococci significantly increased upon (i) lysosomal inhibition by bafilomycin A(1) and (ii) blocking PIKfyve-mediated PtdIns(3,5)P(2) generation by YM201636. In summary, our results provide evidence that the PtdIns(3)P effector function of WIPI-1 is utilized during xenophagy of Staphylococcus aureus. We suggest that invading S. aureus cells become entrapped in autophagosome-like WIPI-1 positive vesicles targeted for lysosomal degradation in nonprofessional host cells.
Collapse
|
47
|
Abstract
Defensins are small, multifunctional cationic peptides. They typically contain six conserved cysteines whose three intramolecular disulfides stabilize a largely β-sheet structure. This review of human α-defensins begins by describing their evolution, including their likely relationship to the Big Defensins of invertebrates, and their kinship to the β-defensin peptides of many if not all vertebrates, and the θ-defensins found in certain non-human primates. We provide a short history of the search for leukocyte-derived microbicidal molecules, emphasizing the roles played by luck (good), preconceived notions (mostly bad), and proper timing (essential). The antimicrobial, antiviral, antitoxic, and binding properties of human α-defensins are summarized. The structural features of α-defensins are described extensively and their functional contributions are assessed. The properties of HD6, an enigmatic Paneth cell α-defensin, are contrasted with those of the four myeloid α-defensins (HNP1-4) and of HD5, the other α-defensin of human Paneth cells. The review ends with a decalogue that may assist researchers or students interested in α-defensins and related aspects of neutrophil function.
Collapse
Affiliation(s)
- Robert I Lehrer
- Department of Medicine and Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1688, USA.
| | | |
Collapse
|
48
|
Passariello C, Puttini M, Iebba V, Pera P, Gigola P. Influence of oral conditions on colonization by highly toxigenic Staphylococcus aureus strains. Oral Dis 2012; 18:402-9. [PMID: 22221343 DOI: 10.1111/j.1601-0825.2011.01889.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVES As the oral cavity is regarded as a relevant site for Staphylococcus aureus colonization and interhuman transmission, this study aimed to investigate whether different oral conditions influence the rates of S. aureus oral carriage and genetic characters of S. aureus isolates. SUBJECTS AND METHODS Staphylococcus aureus was searched in samples collected from cheek, gingival margin, and anterior nares of 45 healthy subjects, 27 periodontitis affected subjects, and 29 subjects with fixed prosthetic restorations. Isolates were screened for 17 genetic determinants, and Partial Least Square Discriminant Analysis was performed to evaluate whether specific characters correlated with oral condition or site of isolation. RESULTS The three subject groups showed comparable nasal carriage rates but, both the periodontitis and prosthetic restoration groups showed significantly higher oral carriage rates, as compared to healthy subjects (P = 0.01 and 0.02, respectively). Moreover, periodontitis affected subjects hosted strains possessing a distinct genotypic and phenotypic background, characterized by the presence of a larger number of exotoxins encoding genes. CONCLUSIONS These data confirm that the oral cavity is an important site of S. aureus colonization and demonstrate that conditions modifying the oral environment, as the presence of periodontitis and of fixed prosthetic restorations, promote S. aureus carriage and may favor the spread of more pathogenic strains.
Collapse
Affiliation(s)
- C Passariello
- Dipartimento di Sanità Pubblica e Malattie Infettive, Università"La Sapienza", Rome, Italy.
| | | | | | | | | |
Collapse
|
49
|
The polyketide Pks1 contributes to biofilm formation in Mycobacterium tuberculosis. J Bacteriol 2011; 194:715-21. [PMID: 22123254 DOI: 10.1128/jb.06304-11] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infections caused by biofilms are abundant and highly persistent, displaying phenotypic resistance to high concentrations of antimicrobials and modulating host immune systems. Tuberculosis (TB), caused by Mycobacterium tuberculosis, shares these qualities with biofilm infections. To identify genetic determinants of biofilm formation in M. tuberculosis, we performed a small-scale transposon screen using an in vitro pellicle biofilm assay. We identified five M. tuberculosis mutants that were reproducibly attenuated for biofilm production relative to that of the parent strain H37Rv. One of the most attenuated mutants is interrupted in pks1, a polyketide synthase gene. When fused with pks15, as in some M. tuberculosis isolates, pks1 contributes to synthesis of the immunomodulatory phenolic glycolipids (PGLs). However, in strains such as H37Rv with split pks15 and pks1 loci, PGL is not produced and pks1 has no previously defined role. We showed that pks1 complementation restores biofilm production independently of the known role of pks1 in PGL synthesis. We also assessed the relationship among biofilm formation, the pks15/1 genotype, and M. tuberculosis phylogeography. A global survey of M. tuberculosis clinical isolates revealed surprising sequence variability in the pks15/1 locus and substantial variation in biofilm phenotypes. Our studies identify novel M. tuberculosis genes that contribute to biofilm production, including pks1. In addition, we find that the ability to make pellicle biofilms is common among M. tuberculosis isolates from throughout the world, suggesting that this trait is relevant to TB propagation or persistence.
Collapse
|
50
|
Suppression of the inflammatory immune response prevents the development of chronic biofilm infection due to methicillin-resistant Staphylococcus aureus. Infect Immun 2011; 79:5010-8. [PMID: 21947772 DOI: 10.1128/iai.05571-11] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is a common cause of prosthetic implant infections, which can become chronic due to the ability of S. aureus to grow as a biofilm. Little is known about adaptive immune responses to these infections in vivo. We hypothesized that S. aureus elicits inflammatory Th1/Th17 responses, associated with biofilm formation, instead of protective Th2/Treg responses. We used an adapted mouse model of biofilm-mediated prosthetic implant infection to determine chronic infection rates, Treg cell frequencies, and local cytokine levels in Th1-biased C57BL/6 and Th2-biased BALB/c mice. All C57BL/6 mice developed chronic S. aureus implant infection at all time points tested. However, over 75% of BALB/c mice spontaneously cleared the infection without adjunctive therapy and demonstrated higher levels of Th2 cytokines and anti-inflammatory Treg cells. When chronic infection rates in mice deficient in the Th2 cytokine interleukin-4 (IL-4) via STAT6 mutation in a BALB/c background were assessed, the mice were unable to clear the S. aureus implant infection. Additionally, BALB/c mice depleted of Treg cells via an anti-CD25 monoclonal antibody (MAb) were also unable to clear the infection. In contrast, the C57BL/6 mice that were susceptible to infection were able to eliminate S. aureus biofilm populations on infected intramedullary pins once the Th1 and Th17 responses were diminished by MAb treatment with anti-IL-12 p40. Together, these results indicate that Th2/Treg responses are mechanisms of protection against chronic S. aureus implant infection, as opposed to Th1/Th17 responses, which may play a role in the development of chronic infection.
Collapse
|