151
|
Ch'ng JH, Muthu M, Chong KKL, Wong JJ, Tan CAZ, Koh ZJS, Lopez D, Matysik A, Nair ZJ, Barkham T, Wang Y, Kline KA. Heme cross-feeding can augment Staphylococcus aureus and Enterococcus faecalis dual species biofilms. THE ISME JOURNAL 2022; 16:2015-2026. [PMID: 35589966 PMCID: PMC9296619 DOI: 10.1038/s41396-022-01248-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 04/18/2022] [Accepted: 04/29/2022] [Indexed: 12/17/2022]
Abstract
The contribution of biofilms to virulence and as a barrier to treatment is well-established for Staphylococcus aureus and Enterococcus faecalis, both nosocomial pathogens frequently isolated from biofilm-associated infections. Despite frequent co-isolation, their interactions in biofilms have not been well-characterized. We report that in combination, these two species can give rise to augmented biofilms biomass that is dependent on the activation of E. faecalis aerobic respiration. In E. faecalis, respiration requires both exogenous heme to activate the cydAB-encoded heme-dependent cytochrome bd, and the availability of O2. We determined that the ABC transporter encoded by cydDC contributes to heme import. In dual species biofilms, S. aureus provides the heme to activate E. faecalis respiration. S. aureus mutants deficient in heme biosynthesis were unable to augment biofilms whereas heme alone is sufficient to augment E. faecalis mono-species biofilms. Our results demonstrate that S. aureus-derived heme, likely in the form of released hemoproteins, promotes E. faecalis biofilm formation, and that E. faecalis gelatinase activity facilitates heme extraction from hemoproteins. This interspecies interaction and metabolic cross-feeding may explain the frequent co-occurrence of these microbes in biofilm-associated infections.
Collapse
Affiliation(s)
- Jun-Hong Ch'ng
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore. .,Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Infectious Disease Translational Research Program, National University Health System, Singapore, Singapore. .,Singapore Centre for Environmental Life Sciences Engineering, National University of Singapore, Singapore, Singapore.
| | - Mugil Muthu
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Kelvin K L Chong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore.,Nanyang Technological University Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore, Singapore
| | - Jun Jie Wong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Interdisciplinary Graduate Program, Nanyang Technological University, Singapore, Singapore
| | - Casandra A Z Tan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Interdisciplinary Graduate Program, Nanyang Technological University, Singapore, Singapore
| | - Zachary J S Koh
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Daniel Lopez
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Artur Matysik
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Zeus J Nair
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Timothy Barkham
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.,Department of Laboratory Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Nanyang, Singapore
| | - Kimberly A Kline
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
152
|
Vlaeminck J, Lin Q, Xavier BB, De Backer S, Berkell M, De Greve H, Hernalsteens JP, Kumar-Singh S, Goossens H, Malhotra-Kumar S. The dynamic transcriptome during maturation of biofilms formed by methicillin-resistant Staphylococcus aureus. Front Microbiol 2022; 13:882346. [PMID: 35966712 PMCID: PMC9366926 DOI: 10.3389/fmicb.2022.882346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023] Open
Abstract
BackgroundMethicillin-resistant Staphylococcus aureus (MRSA), a leading cause of chronic infections, forms prolific biofilms which afford an escape route from antibiotic treatment and host immunity. However, MRSA clones are genetically diverse, and mechanisms underlying biofilm formation remain under-studied. Such studies form the basis for developing targeted therapeutics. Here, we studied the temporal changes in the biofilm transcriptome of three pandemic MRSA clones: USA300, HEMRSA-15, and ST239.MethodsBiofilm formation was assessed using a static model with one representative strain per clone. Total RNA was extracted from biofilm and planktonic cultures after 24, 48, and 72 h of growth, followed by rRNA depletion and sequencing (Illumina Inc., San Diego, CA, United States, NextSeq500, v2, 1 × 75 bp). Differentially expressed gene (DEG) analysis between phenotypes and among early (24 h), intermediate (48 h), and late (72 h) stages of biofilms was performed together with in silico co-expression network construction and compared between clones. To understand the influence of SCCmec and ACME on biofilm formation, isogenic mutants containing deletions of the entire elements or of single genes therein were constructed in USA300.ResultsGenes involved in primarily core genome-encoded KEGG pathways (transporters and others) were upregulated in 24-h biofilm culture compared to 24-h planktonic culture. However, the number of affected pathways in the ST239 24 h biofilm (n = 11) was remarkably lower than that in USA300/EMRSA-15 biofilms (USA300: n = 27, HEMRSA-15: n = 58). The clfA gene, which encodes clumping factor A, was the single common DEG identified across the three clones in 24-h biofilm culture (2.2- to 2.66-fold). In intermediate (48 h) and late (72 h) stages of biofilms, decreased expression of central metabolic and fermentative pathways (glycolysis/gluconeogenesis, fatty acid biosynthesis), indicating a shift to anaerobic conditions, was already evident in USA300 and HEMRSA-15 in 48-h biofilm cultures; ST239 showed a similar profile at 72 h. Last, SCCmec+ACME deletion and opp3D disruption negatively affected USA300 biofilm formation.ConclusionOur data show striking differences in gene expression during biofilm formation by three of the most important pandemic MRSA clones, USA300, HEMRSA-15, and ST239. The clfA gene was the only significantly upregulated gene across all three strains in 24-h biofilm cultures and exemplifies an important target to disrupt early biofilms. Furthermore, our data indicate a critical role for arginine catabolism pathways in early biofilm formation.
Collapse
Affiliation(s)
- Jelle Vlaeminck
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Qiang Lin
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Basil Britto Xavier
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sarah De Backer
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Matilda Berkell
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology & Histology, University of Antwerp, Antwerp, Belgium
| | - Henri De Greve
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology & Histology, University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- *Correspondence: Surbhi Malhotra-Kumar,
| |
Collapse
|
153
|
Phenotypic and Genotypic Characterization of Macrolide, Lincosamide and Streptogramin B Resistance among Clinical Methicillin-Resistant Staphylococcus aureus Isolates in Chile. Antibiotics (Basel) 2022; 11:antibiotics11081000. [PMID: 35892390 PMCID: PMC9332560 DOI: 10.3390/antibiotics11081000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Macrolides, lincosamides, and type B streptogramins (MLSB) are important therapeutic options to treat methicillin-resistant Staphylococcus aureus (MRSA) infections; however, resistance to these antibiotics has been emerging. In Chile, data on the MLSB resistance phenotypes are scarce in both community-(CA) and hospital-acquired (HA) MRSA isolates. Antimicrobial susceptibility to MLSB was determined for sixty-eight non-repetitive isolates of each HA-(32) and CA-MRSA (36). Detection of SCCmec elements, ermA, ermB, ermC, and msrA genes was performed by PCR. The predominant clones were SCCmec I-ST5 (HA-MRSA) and type IVc-ST8 (CA-MRSA). Most of the HA-MRSA isolates (97%) showed resistance to clindamycin, erythromycin, azithromycin, and clarithromycin. Among CA-MRSA isolates, 28% were resistant to erythromycin, azithromycin, and 25% to clarithromycin. All isolates were susceptible to linezolid, vancomycin, daptomycin and trimethoprim/sulfamethoxazole, and over 97% to rifampicin. The ermA gene was amplified in 88% of HA-MRSA and 17% of CA-MRSA isolates (p < 0.001). The ermC gene was detected in 6% of HA-SARM and none of CA-SARM isolates, whereas the msrA gene was only amplified in 22% of CA-MRSA (p < 0.005). Our results demonstrate the prevalence of the cMLSB resistance phenotype in all HA-MRSA isolates in Chile, with the ermA being the predominant gene identified among these isolates.
Collapse
|
154
|
Hawkins AN, Licea SJ, Sleeper SA, Swearingen MC. Calcium sulfate beads made with antibacterial essential oil-water emulsions exhibit growth inhibition against Staphylococcus aureus in agar pour plates. PLoS One 2022; 17:e0271209. [PMID: 35802739 PMCID: PMC9269935 DOI: 10.1371/journal.pone.0271209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/24/2022] [Indexed: 11/21/2022] Open
Abstract
Calcium sulfate bone void filler beads are fully absorbable in the body, and are often used in complicated orthopedic infection cases to release a relatively high dose of antibiotics locally to the body site over time. However, the antibiotic resistance crisis and/or inability to treat chronic biofilm infections remains to be a formidable and increasing health threat. In this report, we tested the hypothesis that plant essential oils (PEOs) with anti-staphylococcal qualities could inhibit the growth of Staphylococcus aureus (a major etiological agent of periprosthetic joint infection) in agar pour plates when infused in calcium sulfate beads. To begin, we conducted a screen of 57 single plant PEOs for anti-staphylococcal activity via disk diffusions assays. We observed that 55/57 of the PEOs had significant growth inhibitory activity compared to the null hypothesis, and 41/57 PEOs exhibited activity similar-to-or-higher-than a vancomycin minimum inhibitory control. When PEOs were infused in beads, we observed that 17/57 PEOs tested exhibited significant bacterial growth inhibition when encased in S. aureus-seeded agar compared to a null hypothesis of six millimeters (bead size). However, none of the PEO-beads had activity similar to a vancomycin bead control made according to a clinically relevant formula. To the best of our knowledge, this is the first report and screen of PEOs for growth inhibitory activity when infused in lab-made calcium sulfate beads. These data indicate that antibacterial PEOs warrant further investigations, and may be useful in developing new treatment strategies for periprosthetic joint infection.
Collapse
Affiliation(s)
- Allison N. Hawkins
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, Florida, United States of America
| | - Sara J. Licea
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, Florida, United States of America
| | - Sierra A. Sleeper
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, Florida, United States of America
| | - Matthew C. Swearingen
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, Florida, United States of America
- * E-mail:
| |
Collapse
|
155
|
The Antibacterial Activity Mode of Action of Plantaricin YKX against Staphylococcus aureus. Molecules 2022; 27:molecules27134280. [PMID: 35807524 PMCID: PMC9268016 DOI: 10.3390/molecules27134280] [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: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 02/01/2023] Open
Abstract
We aimed to evaluate the inhibitory effect and mechanism of plantaricin YKX on S. aureus. The mode of action of plantaricin YKX against the cells of S. aureus indicated that plantaricin YKX was able to cause the leakage of cellular content and damage the structure of the cell membranes. Additionally, plantaricin YKX was also able to inhibit the formation of S. aureus biofilms. As the concentration of plantaricin YKX reached 3/4 MIC, the percentage of biofilm formation inhibition was over 50%. Fluorescent dye labeling combined with fluorescence microscopy confirmed the results. Finally, the effect of plantaricin YKX on the AI-2/LuxS QS system was investigated. Molecular docking predicted that the binding energy of AI-2 and plantaricin YKX was −4.7 kcal/mol and the binding energy of bacteriocin and luxS protein was −183.701 kcal/mol. The expression of the luxS gene increased significantly after being cocultured with plantaricin YKX, suggesting that plantaricin YKX can affect the QS system of S. aureus.
Collapse
|
156
|
Saygin H, Baysal A. Single and combined effects of antibiotics and nanoplastics from surgical masks and plastic bottles on pathogens. Comp Biochem Physiol C Toxicol Pharmacol 2022; 257:109340. [PMID: 35381365 DOI: 10.1016/j.cbpc.2022.109340] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Over the last decade, pollution of plastics and antibiotics has increased in its threat to the environment and human health. However, very limited information is available concerning impact of co-presence of plastics and antibiotics on environment and human health. Moreover, the potential ingestion and inhalation of nano(micro)plastics due to the disposable materials has dramatically increased. With the outbreak and spread of the COVID-19 in the world, disposable surgical masks and plastic bottles have been widely used by the public, and their rapid use and improper dispensing can cause to increase plastic pollution risk on human. However, impacts of co-presence of nano(micro)plastics and antibiotics on pathogens have yet been demonstrated. Therefore, this study aims to investigate the impact the individual and combined influences of nano-sized plastics (surgical mask and plastic bottles) and antibiotics (amoxicillin and spiramycin) towards the main susceptible bacterium (Staphylococcus epidermidis, Staphylococcus aureus, Bacillus subtilis, Escherichia coli, and Pseudomonas aeruginosa) by microbial activity, biofilm formation and their biochemical characteristics. The results showed that antimicrobial efficiencies of the tested antibiotics were reduced (approximately 10-98%) with the plastics. Moreover, the biochemical pathways of the microbial activity changed by the plastics entrance. Polymer structure and sorption play the role on the reduction in the inhibition of pathogens. In the meantime, the biofilm formation changed and characteristic of the extracellular polymeric substance with the co-presence of plastics and antibiotics mostly depended on the polymer structure, exposure time and sorption.
Collapse
Affiliation(s)
- Hasan Saygin
- Application and Research Center for Advanced Studies, T. C. Istanbul Aydin University, Sefakoy Kucukcekmece, 34295 Istanbul, Turkey
| | - Asli Baysal
- Health Services Vocational School of Higher Education, T. C. Istanbul Aydin University, Sefakoy Kucukcekmece, 34295 Istanbul, Turkey.
| |
Collapse
|
157
|
Asharaf S, Chakraborty K. Pharmacological potential of seaweed-associated heterotrophic Firmicutes. Lett Appl Microbiol 2022; 75:1042-1054. [PMID: 35771159 DOI: 10.1111/lam.13780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/11/2022] [Accepted: 06/21/2022] [Indexed: 11/27/2022]
Abstract
Seaweed-associated bacterial symbionts are sources of potential pharmacological properties. The present study resulted in the culture-dependent isolation of bioactive heterotrophs belonging to the bacterial phylum Firmicutes, which were dominated more than 30% of the 127 cultivable isolates, among which 23 of them showed potential antimicrobial activities against a wide range of pathogens. The symbionts isolated from the seaweed Sargassum wightii showed significant bioactivity. Those were characterised as Bacillus safensis MTCC13040, B. valismortis MTCC13041, B. velezensis MTCC13044, B. methylotrophicus MTCC13042, Oceanobacillus profundus MTCC13045, B. tequilensis MTCC13043, and B. altitudinis MTCC13046. The organic extracts of the studied isolates showed potential antimicrobial properties against methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococci (minimum inhibitory concentration 6.25-12.5 μg ml-1 ). The organic extract of B. altitudinis MTCC13046 displayed significantly greater radical quenching ability (IC90 133 μg ml-1 , p < 0.05) other than attenuating hydroxymethyl glutaryl coenzyme A reductase (IC90 10.21 μg ml-1 , p < 0.05) and angiotensin converting enzyme-1 (IC90 498 μg ml-1 , p < 0.05) relative to other studied heterotrophs. The organic extract of B. tequilensis MTCC13043 displayed significantly greater attenuation potential against pro-inflammatory 5-lipooxygenase (IC90 5.94 μg ml-1 , p < 0.05) and dipeptidyl peptidase-4 (IC90 271 μg ml-1 , p < 0.05). The seaweed-associated B. altitudinis MTCC13046 and B. tequilensis MTCC13043 could be used to develop promising pharmacological leads.
Collapse
Affiliation(s)
- Sumayya Asharaf
- Marine Biotechnology Division, Central Marine Fisheries Research Institute, Ernakulam North, P.B. No, 1603, Cochin, -682018, Kerala State, India.,Faculty of Marine Sciences, Lakeside Campus, Cochin University of Science and Technology, Cochin, Kerala State, India
| | - Kajal Chakraborty
- Marine Biotechnology Division, Central Marine Fisheries Research Institute, Ernakulam North, P.B. No, 1603, Cochin, -682018, Kerala State, India
| |
Collapse
|
158
|
AL-Dujaily AH, Mahmood AK. Evaluation of Antibacterial and Antibiofilm Activity of Biogenic Silver Nanoparticles and Gentamicin Against Staphylococcus aureus Isolated from Caprine Mastitis. THE IRAQI JOURNAL OF VETERINARY MEDICINE 2022. [DOI: 10.30539/ijvm.v46i1.1309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The goal of this study was to assess the antibacterial efficiency of biogenic silver nanoparticles (AgNPs) and gentamicin against Staphylococcus aureus that can form biofilms. The characterization of AgNPs was confirmed by the scanning electron microscope (SEM) which was spherical and homogenous in form, with a diameter between 25 and 45 nm. The X-ray diffraction (XRD) presented the size of AgNPs to be 50 nm. Energy dispersive spectroscopy (EDS) was used to examine the presence of elemental silver. The three-dimensional structure of silver nanoparticles was discovered using an atomic force microscope (AFM), with a diameter of 47.18 nm on average. The minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC) of AgNPs and gentamicin against S. aureus isolated from caprine mastitis were determined using the microdilution assay. The checkerboard microdilution technique was utilized to inspect the synergistic antibacterial activity of AgNPs with gentamicin utilizing the fractional inhibitory concentration index (FICI). The antibiofilm capability of AgNPs was also investigated. The results indicate that AgNPs generated by biosynthesis are antibacterial against S. aureus. Moreover, AgNPs and gentamicin exhibit synergistic action. The study's findings suggest that biogenic AgNPs may act as anti-biofilm agents and treat mastitis caused by S. aureus. In conclusions biosynthesized AgNPs exhibit strong antibacterial and antibiofilm effectiveness and synergistic activity when combined with gentamicin.
Collapse
|
159
|
Jin Y, Zhao B, Guo W, Li Y, Min J, Miao W. Penetration and photodynamic ablation of drug-resistant biofilm by cationic Iron oxide nanoparticles. J Control Release 2022; 348:911-923. [PMID: 35760234 DOI: 10.1016/j.jconrel.2022.06.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 10/17/2022]
Abstract
As we step into the post-antibiotic era, the accelerated emergence of antibiotic-resistant pathogenic bacteria poses an increasingly serious threat to public health. The formation of antibiotic-resistant biofilms further challenges currently available drugs and treatment options, calling for novel strategies for effective ablation of such biofilm with minimal concern on safety and development of resistance. Herein, we report a novel type of photodynamic nanoagent, composed of chlorin e6 (Ce6)-loaded water-soluble chitosan-coated iron oxide nanoparticles (named Ce6@WCS-IONP), for drug-resistant bacteria killing and biofilm eradication. The fabricated Ce6@WCS-IONP has negligible toxicity to mammalian cells and exhibited equivalent singlet oxygen generation capacity to free Ce6; however, its association with methicillin-resistant Staphylococcus aureus (MRSA) was greatly enhanced, as evidenced by flow cytometry analysis and transmission electron microscope. In vitro studies verified that Ce6@WCS-IONP has superior photodynamic bactericidal effect against planktonic MRSA. Furthermore, with the aid of the cationic nature and small size, Ce6@WCS-IONP could effectively penetrate into MRSA biofilm, revealed by 3D fluorescence imaging. Both biomass analysis and viable bacteria counting demonstrated that Ce6@WCS-IONP showed potent biofilm ablation efficacy, averagely 7.1 log unit lower than that in free Ce6 group upon identical light irradiation. In addition, local treatment of MRSA-infected mice with Ce6@WCS-IONP plus light irradiation resulted in significant antibacterial and wound healing effect, accompanied by good biocompatibility in vivo. Collectively, photosensitizer-loaded cationic IONP with effective biofilm penetration and photodynamic eradication potential might be a promising nano platform in fighting against antibiotic-resistant microbial pathogen and biofilm.
Collapse
Affiliation(s)
- Yangye Jin
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, PR China
| | - Binbing Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, PR China
| | - Wenjing Guo
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, PR China
| | - Yuanyuan Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, PR China
| | - Juncheng Min
- College of Overseas Education, Nanjing Tech University, Nanjing 211816, PR China
| | - Wenjun Miao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, PR China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, PR China.
| |
Collapse
|
160
|
Ballah FM, Islam MS, Rana ML, Ferdous FB, Ahmed R, Pramanik PK, Karmoker J, Ievy S, Sobur MA, Siddique MP, Khatun MM, Rahman M, Rahman MT. Phenotypic and Genotypic Detection of Biofilm-Forming Staphylococcus aureus from Different Food Sources in Bangladesh. BIOLOGY 2022; 11:biology11070949. [PMID: 36101330 PMCID: PMC9311614 DOI: 10.3390/biology11070949] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/30/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022]
Abstract
Staphylococcus aureus is a major foodborne pathogen. The ability of S. aureus to produce biofilm is a significant virulence factor, triggering its persistence in hostile environments. In this study, we screened a total of 420 different food samples and human hand swabs to detect S. aureus and to determine their biofilm formation ability. Samples analyzed were meat, milk, eggs, fish, fast foods, and hand swabs. S. aureus were detected by culturing, staining, biochemical, and PCR. Biofilm formation ability was determined by Congo Red Agar (CRA) plate and Crystal Violet Microtiter Plate (CVMP) tests. The icaA, icaB, icaC, icaD, and bap genes involved in the synthesis of biofilm-forming intracellular adhesion compounds were detected by PCR. About 23.81% (100/420; 95% CI: 14.17−29.98%) of the samples harbored S. aureus, as revealed by detection of the nuc gene. The CRA plate test revealed 20% of S. aureus isolates as strong biofilm producers and 69% and 11% as intermediate and non-biofilm producers, respectively. By the CVMP staining method, 20%, 77%, and 3% of the isolates were found to be strong, intermediate, and non-biofilm producers. Furthermore, 21% of S. aureus isolates carried at least one biofilm-forming gene, where icaA, icaB, icaC, icaD, and bap genes were detected in 15%, 20%, 7%, 20%, and 10% of the S. aureus isolates, respectively. Bivariate analysis showed highly significant correlations (p < 0.001) between any of the two adhesion genes of S. aureus isolates. To the best of our knowledge, this is the first study in Bangladesh describing the detection of biofilm-forming S. aureus from foods and hand swabs using molecular-based evidence. Our findings suggest that food samples should be deemed a potential reservoir of biofilm-forming S. aureus, which indicates a potential public health significance.
Collapse
|
161
|
Deepika G, Subbarayadu S, Chaudhary A, Sarma PVGK. Dibenzyl (benzo [d] thiazol-2-yl (hydroxy) methyl) phosphonate (DBTMP) showing anti-S. aureus and anti-biofilm properties by elevating activities of serine protease (SspA) and cysteine protease staphopain B (SspB). Arch Microbiol 2022; 204:397. [PMID: 35708833 DOI: 10.1007/s00203-022-02974-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Staphylococcus aureus biofilms are the pathogenic factor in the spread of infection and are more pronounced in multidrug-resistant strains of S. aureus, where high expression of proteases is observed. Among various proteases, Serine protease (SspA) and cysteine protease Staphopain B (SspB) are known to play a key role in the biofilm formation and removal of biofilms. In earlier studies, we have reported Dibenzyl (benzo [d] thiazol-2-yl (hydroxy) methyl) phosphonate (DBTMP) exhibits anti-S. aureus and anti-biofilm properties by elevating the expression of the protease. In this study, the effect of DBTMP on the activities of SspA, and SspB of S. aureus was evaluated. The SspA and SspB genes of S. aureus ATCC12600 were sequenced (Genbank accession numbers: MZ456982 and MW574006). In S. aureus active SspA is formed by proteolytic cleavage of immature SspA, to get this mature SspA (mSspA), we have PCR amplified the mSspA sequence from the SspA gene. The mSspA and SspB genes were cloned, expressed, and characterized. The pure recombinant proteins rSspB and rmSspA exhibited a single band in SDS-PAGE with a molecular weight of 40 and 30 KD, respectively. The activities of rmSspA and rSspB are 32.33 and 35.45 Units/mL correspondingly. DBTMP elevated the activities of rmSspA and rSspB by docking with respective enzymes. This compound disrupted the biofilms formed by the multidrug-resistant strains of S. aureus and further prevented biofilm formation. These findings explain that DBTMP possesses anti-S. aureus and anti-biofilm features.
Collapse
Affiliation(s)
- G Deepika
- Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, 517507, India
| | - S Subbarayadu
- Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, 517507, India
| | - Abhijit Chaudhary
- Department of Microbiology, Sri Padmavati Medical College (Women), SVIMS, Tirupati, Andhra Pradesh, 517507, India
| | - P V G K Sarma
- Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, 517507, India.
| |
Collapse
|
162
|
Wang M, Buist G, van Dijl JM. Staphylococcus aureus cell wall maintenance - the multifaceted roles of peptidoglycan hydrolases in bacterial growth, fitness, and virulence. FEMS Microbiol Rev 2022; 46:6604383. [PMID: 35675307 PMCID: PMC9616470 DOI: 10.1093/femsre/fuac025] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is an important human and livestock pathogen that is well-protected against environmental insults by a thick cell wall. Accordingly, the wall is a major target of present-day antimicrobial therapy. Unfortunately, S. aureus has mastered the art of antimicrobial resistance, as underscored by the global spread of methicillin-resistant S. aureus (MRSA). The major cell wall component is peptidoglycan. Importantly, the peptidoglycan network is not only vital for cell wall function, but it also represents a bacterial Achilles' heel. In particular, this network is continuously opened by no less than 18 different peptidoglycan hydrolases (PGHs) encoded by the S. aureus core genome, which facilitate bacterial growth and division. This focuses attention on the specific functions executed by these enzymes, their subcellular localization, their control at the transcriptional and post-transcriptional levels, their contributions to staphylococcal virulence and their overall importance in bacterial homeostasis. As highlighted in the present review, our understanding of the different aspects of PGH function in S. aureus has been substantially increased over recent years. This is important because it opens up new possibilities to exploit PGHs as innovative targets for next-generation antimicrobials, passive or active immunization strategies, or even to engineer them into effective antimicrobial agents.
Collapse
Affiliation(s)
- Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | | | - Jan Maarten van Dijl
- Corresponding author: Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. box 30001, HPC EB80, 9700 RB Groningen, the Netherlands, Tel. +31-50-3615187; Fax. +31-50-3619105; E-mail:
| |
Collapse
|
163
|
Weaver AJ, Borgogna TR, O’Shea-Stone G, Peters TR, Copié V, Voyich J, Teintze M. 18β-Glycyrrhetinic Acid Induces Metabolic Changes and Reduces Staphylococcus aureus Bacterial Cell-to-Cell Interactions. Antibiotics (Basel) 2022; 11:antibiotics11060781. [PMID: 35740189 PMCID: PMC9220049 DOI: 10.3390/antibiotics11060781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 12/04/2022] Open
Abstract
The rise in bacterial resistance to common antibiotics has raised an increased need for alternative treatment strategies. The natural antibacterial product, 18β-glycyrrhetinic acid (GRA) has shown efficacy against community-associated methicillin-resistant Staphylococcus aureus (MRSA), although its interactions against planktonic and biofilm modes of growth remain poorly understood. This investigation utilized biochemical and metabolic approaches to further elucidate the effects of GRA on MRSA. Prolonged exposure of planktonic MRSA cell cultures to GRA resulted in increased production of staphyloxanthin, a pigment known to exhibit antioxidant and membrane-stabilizing functions. Then, 1D 1H NMR analyses of intracellular metabolite extracts from MRSA treated with GRA revealed significant changes in intracellular polar metabolite profiles, including increased levels of succinate and citrate, and significant reductions in several amino acids, including branch chain amino acids. These changes reflect the MRSA response to GRA exposure, including potentially altering its membrane composition, which consumes branched chain amino acids and leads to significant energy expenditure. Although GRA itself had no significant effect of biofilm viability, it seems to be an effective biofilm disruptor. This may be related to interference with cell–cell aggregation, as treatment of planktonic MRSA cultures with GRA leads to a significant reduction in micro-aggregation. The dispersive nature of GRA on MRSA biofilms may prove valuable for treatment of such infections and could be used to increase susceptibility to complementary antibiotic therapeutics.
Collapse
Affiliation(s)
- Alan J. Weaver
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT 59717, USA; (A.J.W.J.); (G.O.-S.); (T.R.P.)
| | - Timothy R. Borgogna
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717, USA;
| | - Galen O’Shea-Stone
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT 59717, USA; (A.J.W.J.); (G.O.-S.); (T.R.P.)
| | - Tami R. Peters
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT 59717, USA; (A.J.W.J.); (G.O.-S.); (T.R.P.)
| | - Valérie Copié
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT 59717, USA; (A.J.W.J.); (G.O.-S.); (T.R.P.)
- Correspondence: (V.C.); (J.V.); (M.T.); Tel.: +406-994-7244 (V.C.); +406-994-7184 (J.V.); +406-994-6515 (M.T.)
| | - Jovanka Voyich
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717, USA;
- Correspondence: (V.C.); (J.V.); (M.T.); Tel.: +406-994-7244 (V.C.); +406-994-7184 (J.V.); +406-994-6515 (M.T.)
| | - Martin Teintze
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT 59717, USA; (A.J.W.J.); (G.O.-S.); (T.R.P.)
- Correspondence: (V.C.); (J.V.); (M.T.); Tel.: +406-994-7244 (V.C.); +406-994-7184 (J.V.); +406-994-6515 (M.T.)
| |
Collapse
|
164
|
Dollery SJ, Harro JM, Wiggins TJ, Wille BP, Kim PC, Tobin JK, Bushnell RV, Tasker NJPER, MacLeod DA, Tobin GJ. Select Whole-Cell Biofilm-Based Immunogens Protect against a Virulent Staphylococcus Isolate in a Stringent Implant Model of Infection. Vaccines (Basel) 2022; 10:833. [PMID: 35746441 PMCID: PMC9231243 DOI: 10.3390/vaccines10060833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Many microbes of concern to human health remain without vaccines. We have developed a whole-microbe inactivation technology that enables us to rapidly inactivate large quantities of a pathogen while retaining epitopes that were destroyed by previous inactivation methods. The method that we call UVC-MDP inactivation can be used to make whole-cell vaccines with increased potency. We and others are exploring the possibility of using improved irradiation-inactivation technologies to develop whole-cell vaccines for numerous antibiotic-resistant microbes. Here, we apply UVC-MDP to produce candidate MRSA vaccines which we test in a stringent tibia implant model of infection challenged with a virulent MSRA strain. We report high levels of clearance in the model and observe a pattern of protection that correlates with the immunogen protein profile used for vaccination.
Collapse
Affiliation(s)
- Stephen J. Dollery
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Janette M. Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - Taralyn J. Wiggins
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Brendan P. Wille
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - Peter C. Kim
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - John K. Tobin
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Ruth V. Bushnell
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Naomi J. P. E. R. Tasker
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - David A. MacLeod
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Gregory J. Tobin
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| |
Collapse
|
165
|
Yin X, Fang Z, Fang Y, Zhu L, Pang J, Liu T, Zhao Z, Zhao J. Antimicrobial Photodynamic Therapy Involving a Novel Photosensitizer Combined With an Antibiotic in the Treatment of Rabbit Tibial Osteomyelitis Caused by Drug-Resistant Bacteria. Front Microbiol 2022; 13:876166. [PMID: 35531297 PMCID: PMC9073078 DOI: 10.3389/fmicb.2022.876166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
Osteomyelitis is deep tissue inflammation caused by bacterial infection. If such an infection persists, it can lead to dissolution and necrosis of the bone tissue. As a result of the extensive use of antibiotics, drug-resistant bacteria are an increasingly common cause of osteomyelitis, limiting the treatment options available to surgeons. Photodynamic antibacterial chemotherapy has attracted increasing attention as a potential alternative treatment. Its advantages are a broad antibacterial spectrum, lack of drug resistance, and lack of toxic side effects. In this study, we explored the impact of the new photosensitizer LD4 in photodynamic antimicrobial chemotherapy (PACT), both alone and in combination with an antibiotic, on osteomyelitis. A rabbit tibial osteomyelitis model was employed and microbiological, histological, and radiological studies were performed. New Zealand white rabbits (n = 36) were randomly divided into a control group, antibiotic group, PACT group and PACT + antibiotic group for treatment. In microbiological analysis, a reduction in bacterial numbers of more than 99.9% was recorded in the PACT group and the PACT + antibiotic group 5 weeks after treatment (p < 0.01). In histological analysis, repair of the damaged bone tissue was observed in the PACT group, and bone repair in the PACT + antibiotic group was even more significant. In radiological analysis, the X-ray Norden score showed that the severity of bone tissue defects or destruction followed the pattern: PACT + antibiotic group < PACT group < antibiotic group < control group.
Collapse
Affiliation(s)
- Xiujuan Yin
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Ziyuan Fang
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Yan Fang
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Lin Zhu
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Jinwen Pang
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Tianjun Liu
- Tianjin Key Laboratory of Biomedical Material, Institute of Biomedical Engineering, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Zhanjuan Zhao
- School of Basic Medicine, Hebei University, Baoding, China
| | - Jianxi Zhao
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
166
|
Ganesh P, Veena K, Senthil R, Iswamy K, Ponmalar EM, Mariappan V, Girija ASS, Vadivelu J, Nagarajan S, Challabathula D, Shankar EM. Biofilm-Associated Agr and Sar Quorum Sensing Systems of Staphylococcus aureus Are Inhibited by 3-Hydroxybenzoic Acid Derived from Illicium verum. ACS OMEGA 2022; 7:14653-14665. [PMID: 35557687 PMCID: PMC9088959 DOI: 10.1021/acsomega.1c07178] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/31/2022] [Indexed: 05/10/2023]
Abstract
Biofilm-producing Staphylococcus aureus (S. aureus) is less sensitive to conventional antibiotics than free-living planktonic cells. Here, we evaluated the antibiofilm activity of Illicium verum (I. verum) and one of its constituent compounds 3-hydroxybenzoic acid (3-HBA) against multi-drug-resistant S. aureus. We performed gas chromatography-mass spectroscopy (GC-MS) to identify the major constituents in the methanolic extract of I. verum. Ligand-receptor interactions were studied by molecular docking, and in vitro investigations were performed using crystal violet assay, spreading assay, hemolysis, proteolytic activity, and growth curve analysis. The methanolic extract of I. verum inhibited S. aureus at 4.8 mg/mL, and GC-MS analysis revealed anethole, m-methoxybenzaldehyde, and 3-HBA as the major constituents. Molecular docking attributed the antibiofilm activity to an active ligand present in 3-HBA, which strongly interacted with the active site residues of AgrA and SarA of S. aureus. At a subinhibitory concentration of 2.4 mg/mL, the extract showed biofilm inhibition. Similarly, 3-HBA inhibited biofilm activity at 25 μg/mL (90.34%), 12.5 μg/mL (77.21%), and 6.25 μg/mL (62.69%) concentrations. Marked attrition in bacterial spreading was observed at 2.4 mg/mL (crude extract) and 25 μg/mL (3-HBA) concentrations. The methanol extract of I. verum and 3-HBA markedly inhibited β-hemolytic and proteolytic activities of S. aureus. At the lowest concentration, the I. verum extract (2.4 mg/mL) and 3-HBA (25 μg/mL) did not inhibit bacterial growth. Optical microscopy and SEM analysis confirmed that I. verum and 3-HBA significantly reduced biofilm dispersion without disturbing bacterial growth. Together, we found that the antibiofilm activity of I. verum and 3-HBA strongly targeted the Agr and Sar systems of S. aureus.
Collapse
Affiliation(s)
- Pitchaipillai
Sankar Ganesh
- Department
of Microbiology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Velappanchavadi, PH Road, Chennai 600077, Tamilnadu, India
| | - Krishnamurthy Veena
- Infection
Biology, Department of Life Sciences, Central
University of Tamil Nadu, Neelakudi, Tiruvarur 610005, Tamilnadu, India
| | - Renganathan Senthil
- Department
of Bioinformatics, Marudupandiyar College, Vallam, Thanjavur 613403, Tamilnadu, India
| | - Koneti Iswamy
- Infection
Biology, Department of Life Sciences, Central
University of Tamil Nadu, Neelakudi, Tiruvarur 610005, Tamilnadu, India
| | - Esaki Muthu Ponmalar
- Sri
Sairam Siddha Medical College and Research Centre, West Tambaram, Chennai 600044, Tamilnadu, India
| | - Vanitha Mariappan
- Center
for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - A. S. Smiline Girija
- Department
of Microbiology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Velappanchavadi, PH Road, Chennai 600077, Tamilnadu, India
| | - Jamuna Vadivelu
- Department
of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 50603, Malaysia
| | - Samuthira Nagarajan
- Department
of Chemistry, Central University of Tamil
Nadu, Neelakudi, Tiruvarur 610005, Tamil Nadu, India
| | - Dinakar Challabathula
- Department
of Life Sciences, Central University of
Tamil Nadu, Neelakudi, Tiruvarur 610005, Tamil Nadu, India
| | - Esaki Muthu Shankar
- Infection
Biology, Department of Life Sciences, Central
University of Tamil Nadu, Neelakudi, Tiruvarur 610005, Tamilnadu, India
| |
Collapse
|
167
|
Chitosan-gum arabic embedded alizarin nanocarriers inhibit biofilm formation of multispecies microorganisms. Carbohydr Polym 2022; 284:118959. [DOI: 10.1016/j.carbpol.2021.118959] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/17/2021] [Accepted: 11/28/2021] [Indexed: 01/09/2023]
|
168
|
Sinsinwar S, Jayaraman A, Mahapatra SK, Vellingiri V. Anti-virulence properties of catechin-in-cyclodextrin-in-phospholipid liposome through down-regulation of gene expression in MRSA strains. Microb Pathog 2022; 167:105585. [DOI: 10.1016/j.micpath.2022.105585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022]
|
169
|
Fan Q, Yuan Y, Zhang T, Song W, Sheng Q, Yue T. Inhibitory effects of lactobionic acid on Vibrio parahaemolyticus planktonic cells and biofilms. Food Microbiol 2022; 103:103963. [DOI: 10.1016/j.fm.2021.103963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022]
|
170
|
Antibiotic-loaded lipid-based nanocarrier: a promising strategy to overcome bacterial infection. Int J Pharm 2022; 621:121782. [PMID: 35489605 DOI: 10.1016/j.ijpharm.2022.121782] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 12/18/2022]
Abstract
According to the World Health Organization (WHO) and Centers for Disease Control and Prevention (CDC), bacterial infections are one of the greatest threats to global health, food production, and life expectancy. In this sense, the development of innovative formulations aiming at greater therapeutic efficacy, safety, and shorter treatment duration compared to conventional products is urgently needed. Lipid-based nanocarriers (LBNs) have demonstrated the potential to enhance the effectiveness of available antibiotics. Among them, liposome, nanoemulsion, solid lipid nanoparticle (SLN), and nanostructured lipid carrier (NLC) are the most promising due to their solid technical background for laboratory and industrial production. This review describes recent advances in developing antibiotic-loaded LBNs against susceptible and resistant bacterial strains and biofilm. LBNs revealed to be a promising alternative to deliver antibiotics due to their superior characteristics compared to conventional preparations, including their modified drug release, improved bioavailability, drug protection against chemical or enzymatic degradation, greater drug loading capacity, and biocompatibility. Antibiotic-loaded LBNs can improve current clinical drug therapy, bring innovative products and rescue discarded antibiotics. Thus, antibiotic-loaded LBNs have potential to open a window of opportunities to continue saving millions of lives and prevent the devastating impact of bacterial infection.
Collapse
|
171
|
Zank A, Schulte L, Brandon X, Carstensen L, Wescott A, Schwan WR. Mutations of the brpR and brpS genes affect biofilm formation in Staphylococcus aureus. World J Clin Infect Dis 2022; 12:20-32. [DOI: 10.5495/wjcid.v12.i1.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/03/2021] [Accepted: 02/13/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In the United States, Staphylococcus aureus (S. aureus) kills tens of thousands of individuals each year and the formation of a biofilm contributes to lethality. Biofilm-associated infections are hard to treat once the biofilm has formed. A new stilbene drug, labeled SK-03-92, was shown to kill S. aureus and affected transcription of two genes tied to a putative two-component system (TCS) we have named brpR (biofilm regulating protein regulator) and brpS (biofilm regulating protein sensor).
AIM To determine if BrpR and BrpS regulate biofilm formation, brpR and brpS mutants were assessed using biofilm assays compared to wild-type S. aureus.
METHODS A combination of biofilm and quantitative real-time-polymerase chain reaction assays were used. In addition, bioinformatic software tools were also utilized.
RESULTS Significantly more biofilm was created in the brpR and brpS mutants vs wild-type cells. Quantitative real-time polymerase chain reactions showed the brpS mutant had differences in transcription of biofilm associated genes that were eight-fold higher for srtA, two-fold lower for lrgA, and 1.6-fold higher for cidA compared to wild-type. Bioinformatic analysis demonstrated that the S. aureus brpR/brpS TCS had homology to streptococcal late-stage competence proteins involved in cell-death, increased biofilm production, and the development of persister cells.
CONCLUSION Our study suggests that brpR/brpS is a TCS that may repress S. aureus biofilm production and be linked to late-stage competence in S. aureus.
Collapse
Affiliation(s)
- Allison Zank
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Lillian Schulte
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Xavier Brandon
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Lauren Carstensen
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - Amy Wescott
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| | - William R Schwan
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, United States
| |
Collapse
|
172
|
Manandhar S, Singh A, Varma A, Pandey S, Shrivastava N. High level of persister frequency in clinical staphylococcal isolates. BMC Microbiol 2022; 22:109. [PMID: 35448965 PMCID: PMC10124895 DOI: 10.1186/s12866-022-02529-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 04/12/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is a notorious human pathogen that causes often lethal systemic conditions that are mostly medical device associated biofilm infections. Similarly, coagulase negative staphylococci are emerging as leading pathogen for nosocomial infections owing to their ability to form biofilm on implanted medical equipment. Chronic in nature, these infections are difficult to treat. Such recalcitrance of these infections is caused mainly due to the presence of persister cells, which exhibit transient yet extreme tolerance to antibiotics. Despite tremendous clinical significance, there is lack of studies on persister cells formation among clinical bacterial isolates. Considering the importance of factors influencing persister formation, in this study, we evaluate the association of antibiotic tolerance with biofilm production, antibiotic stress, growth phase, specimen type, and dependency on staphylococcal species. Biofilm formation was detected among 375 clinical staphylococcal isolates by quantitative tissue culture plate method (TCP) and icaAD genes by genotypic method. The antibiotic susceptibility was determined by Kirby Bauer disc diffusion method while minimum inhibitory concentration values were obtained by agar dilution method. Persister cells were measured in the susceptible staphylococcal isolates in the presence of clinically relevant antibiotics. RESULTS In the study, 161 (43%) S. aureus and 214 (57%) coagulase negative staphylococci (CNS) were isolated from different clinical samples. TCP method detected biofilm production in 84 (52.2%) S. aureus and 90 (42.1%) CNS isolates. The genotypic method detected icaAD genes in 86 (22.9%) isolates. Majority (> 90%) of both the biofilm producers and non-producers were sensitive to chloramphenicol and tetracycline but were resistant to penicillin. Interestingly, all isolates were sensitive to vancomycin irrespective of biofilm production. While high persister frequency was observed among all staphylococci isolates in the stationary growth phase, the persister frequency in exponential growth phase was statistically high among isolates possessing icaAD genes compared to icaAD negative isolates. CONCLUSION The research findings provide strong evidence that the clinical staphylococcal isolates exhibit extreme antibiotic tolerance suggesting their causal link with treatment failures. Understanding the factors influencing the formation and maintenance of persister cells are of utmost important aspect to design therapeutics and control recalcitrant bacterial infections.
Collapse
Affiliation(s)
- Sarita Manandhar
- Tri-Chandra Multiple College, Tribhuvan University, Kathmandu, Nepal.
| | - Anjana Singh
- Central Department of Microbiology, Tribhuvan University, Kathmandu, Nepal
| | - Ajit Varma
- Amity Institute of Microbial Technology, Amity University, Uttar Pradesh, Noida, UP, 201303, India
| | - Shanti Pandey
- The University of Southern Mississippi, Hattiesburg, MS-39406, USA
| | - Neeraj Shrivastava
- Amity Institute of Microbial Technology, Amity University, Uttar Pradesh, Noida, UP, 201303, India
| |
Collapse
|
173
|
Yaacob SN, Wahab RA, Misson M, Sabullah MK, Huyop F, Zin NM. Lactic acid bacteria and their bacteriocins: new potential weapons in the fight against methicillin-resistant Staphylococcus aureus. Future Microbiol 2022; 17:683-699. [PMID: 35414206 DOI: 10.2217/fmb-2021-0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alternative solutions are eminently needed to combat the escalating number of infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Bacteriocins produced by lactic acid bacteria are promising candidates for next-generation antibiotics. Studies have found that these stable and nontoxic ribosomally synthesized antimicrobial peptides exhibit significant potency against other bacteria, including antibiotic-resistant strains. Here the authors review previous studies on bacteriocins that have been effectively employed to manage MRSA infections. The authors' review focuses on the beneficial traits of bacteriocins for further application as templates for the design of novel drugs. Treatments that combine bacteriocins with other antimicrobials to combat pervasive MRSA infections are also highlighted. In short, future studies should focus on the pharmacodynamics and pharmacokinetics of bacteriocins-antimicrobials to understand their interactions, as this aspect would likely determine their efficacy in MRSA inhibition.
Collapse
Affiliation(s)
- Syariffah Ns Yaacob
- Department of Bioscience, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Malaysia
| | - Roswanira A Wahab
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Malaysia
| | - Mailin Misson
- Biotechnology Research Institute, Jalan Universiti Malaysia Sabah, Kota Kinabalu, Sabah, 88400, Malaysia
| | - Mohd K Sabullah
- Faculty of Science and Natural Resources, Jalan Universiti Malaysia Sabah, Kota Kinabalu, Sabah, 88400, Malaysia
| | - Fahrul Huyop
- Department of Bioscience, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Malaysia
| | - Noraziah M Zin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| |
Collapse
|
174
|
Chiba A, Seki M, Suzuki Y, Kinjo Y, Mizunoe Y, Sugimoto S. Staphylococcus aureus utilizes environmental RNA as a building material in specific polysaccharide-dependent biofilms. NPJ Biofilms Microbiomes 2022; 8:17. [PMID: 35379830 PMCID: PMC8980062 DOI: 10.1038/s41522-022-00278-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
Biofilms are surface-bound microbial communities that are typically embedded in a matrix of self-produced extracellular polymeric substances and can cause chronic infections. Extracellular DNA is known to play a crucial role in biofilm development in diverse bacteria; however, the existence and function of RNA are poorly understood. Here, we show that RNA contributes to the structural integrity of biofilms formed by the human pathogen Staphylococcus aureus. RNase A dispersed both fresh and mature biofilms, indicating the importance of RNA at various stages. RNA-sequencing analysis demonstrated that the primary source of RNA in the biofilm matrix was the Brain Heart Infusion medium (>99.32%). RNA purified from the medium promoted biofilm formation. Microscopic and molecular interaction analyses demonstrated that polysaccharides were critical for capturing and stabilizing external RNA in biofilms, which contributes to biofilm organization. These findings provide a basis for exploring the role of externally derived substances in bacterial biofilm organization.
Collapse
|
175
|
Klagisa R, Racenis K, Broks R, Kise L, Kroiča J. Evaluation of Staphylococcus aureus Colonization in Adult Patients Undergoing Tonsillectomy for Recurrent Tonsillitis. Pathogens 2022; 11:pathogens11040427. [PMID: 35456100 PMCID: PMC9029959 DOI: 10.3390/pathogens11040427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Background and objectives: Staphylococcus aureus (S. aureus) is often recovered from the pharynx. However, the role of this pathogen in the etiology of tonsillar inflammation is still unclear and complicated due to frequent carriage of S. aureus. The aim of the study was to evaluate the frequency and the clinical importance of S. aureus colonization and biofilm production ability in patients with recurrent tonsillitis (RT) using patient samples from tonsillar crypts during tonsillectomy, and from the throat, nasal cavity, and armpits after tonsillectomy. Materials and Methods: A case series study was carried out at a tertiary referral center among 16 patients diagnosed with RT who were undergoing tonsillectomy. Samples from tonsillar crypts were obtained during tonsillectomy, and samples from the throat, nasal cavity, and armpit were obtained a year after surgery. An evaluation of S. aureus incidence, biofilm formation, and antibacterial susceptibility was performed. Results: During tonsillectomy, 16 strains of S. aureus were isolated from 16 patients, while 15/16 S. aureus strains were biofilm producers. A year after tonsillectomy, 8 S. aureus strains were isolated from 6 out of 16 patients, while 6/8 S. aureus strains were biofilm producers. After tonsillectomy, 3 patients showed S. aureus in throat culture. Conclusions: In 10/16 cases S. aureus was the causative agent of RT, in 4/16 cases patients had a predisposition to colonization of S. aureus, and in 2/16 cases S. aureus was a part of the patients` oral microbiome. Tonsillectomy results in less frequent isolation of S. aureus strains.
Collapse
Affiliation(s)
- Renata Klagisa
- Department of Otorhinolaryngology, Daugavpils Regional Hospital, LV-5401 Daugavpils, Latvia
- Department of Doctoral Studies, Riga Stradins University, LV-1007 Riga, Latvia;
- Correspondence: ; Tel.: +371-28471191
| | - Karlis Racenis
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia; (K.R.); (R.B.); (J.K.)
- Center of Nephrology, Pauls Stradins Clinical University Hospital, LV-1002 Riga, Latvia
| | - Renars Broks
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia; (K.R.); (R.B.); (J.K.)
| | - Ligija Kise
- Department of Doctoral Studies, Riga Stradins University, LV-1007 Riga, Latvia;
| | - Juta Kroiča
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia; (K.R.); (R.B.); (J.K.)
| |
Collapse
|
176
|
Darvishi S, Tavakoli S, Kharaziha M, Girault HH, Kaminski CF, Mela I. Advances in the Sensing and Treatment of Wound Biofilms. Angew Chem Int Ed Engl 2022; 61:e202112218. [PMID: 34806284 PMCID: PMC9303468 DOI: 10.1002/anie.202112218] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 12/02/2022]
Abstract
Wound biofilms represent a particularly challenging problem in modern medicine. They are increasingly antibiotic resistant and can prevent the healing of chronic wounds. However, current treatment and diagnostic options are hampered by the complexity of the biofilm environment. In this review, we present new chemical avenues in biofilm sensors and new materials to treat wound biofilms, offering promise for better detection, chemical specificity, and biocompatibility. We briefly discuss existing methods for biofilm detection and focus on novel, sensor-based approaches that show promise for early, accurate detection of biofilm formation on wound sites and that can be translated to point-of-care settings. We then discuss technologies inspired by new materials for efficient biofilm eradication. We focus on ultrasound-induced microbubbles and nanomaterials that can both penetrate the biofilm and simultaneously carry active antimicrobials and discuss the benefits of those approaches in comparison to conventional methods.
Collapse
Affiliation(s)
- Sorour Darvishi
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Shima Tavakoli
- Department of Chemistry-Ångstrom LaboratoryUppsala UniversitySE75121UppsalaSweden
| | - Mahshid Kharaziha
- Department of Materials EngineeringIsfahan University of TechnologyIsfahan84156-83111Iran
| | - Hubert H. Girault
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Clemens F. Kaminski
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Ioanna Mela
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
177
|
Darvishi S, Tavakoli S, Kharaziha M, Girault HH, Kaminski CF, Mela I. Advances in the Sensing and Treatment of Wound Biofilms. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202112218. [PMID: 38505642 PMCID: PMC10946914 DOI: 10.1002/ange.202112218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 03/21/2024]
Abstract
Wound biofilms represent a particularly challenging problem in modern medicine. They are increasingly antibiotic resistant and can prevent the healing of chronic wounds. However, current treatment and diagnostic options are hampered by the complexity of the biofilm environment. In this review, we present new chemical avenues in biofilm sensors and new materials to treat wound biofilms, offering promise for better detection, chemical specificity, and biocompatibility. We briefly discuss existing methods for biofilm detection and focus on novel, sensor-based approaches that show promise for early, accurate detection of biofilm formation on wound sites and that can be translated to point-of-care settings. We then discuss technologies inspired by new materials for efficient biofilm eradication. We focus on ultrasound-induced microbubbles and nanomaterials that can both penetrate the biofilm and simultaneously carry active antimicrobials and discuss the benefits of those approaches in comparison to conventional methods.
Collapse
Affiliation(s)
- Sorour Darvishi
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Shima Tavakoli
- Department of Chemistry-Ångstrom LaboratoryUppsala UniversitySE75121UppsalaSweden
| | - Mahshid Kharaziha
- Department of Materials EngineeringIsfahan University of TechnologyIsfahan84156-83111Iran
| | - Hubert H. Girault
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Clemens F. Kaminski
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Ioanna Mela
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
178
|
Use of Photodynamic Therapy Associated with Antimicrobial Peptides for Bacterial Control: A Systematic Review and Meta-Analysis. Int J Mol Sci 2022; 23:ijms23063226. [PMID: 35328647 PMCID: PMC8953507 DOI: 10.3390/ijms23063226] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 12/29/2022] Open
Abstract
Considering the challenges related to antimicrobial resistance, other strategies for controlling infections have been suggested, such as antimicrobial photodynamic therapy (aPDT) and antimicrobial peptides (AMP). This study aims to perform a systematic review and meta-analysis to obtain evidence on the antimicrobial effectiveness of aPDT associated with AMP and establish in vitro knowledge on this topic for further study designs. The PubMed, Scopus, Web of Science, Science Direct, Scielo, and Cochrane Library databases were searched. Two independent and calibrated researchers (Kappa = 0.88) performed all the systematic steps according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). The odds ratio (OR) was used as the effect measure. The Peto method was used to perform the meta-analysis due to the sparse data. Twenty studies were included in the present review. The result was significant (OR = 0.14/p = 0.0235/I-squared = 0%), showing better outcomes of aPDT associated with peptides than those of aPDT alone for controlling the microbial load. Only 20% of the studies included evaluated this approach in a biofilm culture. Combined treatment with aPDT and AMP highly increased the ability of microbial reduction of Gram-positive and Gram-negative bacteria. However, additional blind studies are required to evaluate the efficacy of this therapy on microbial biofilms.
Collapse
|
179
|
|
180
|
Antibacterial Activity of Isobavachalcone (IBC) Is Associated with Membrane Disruption. MEMBRANES 2022; 12:membranes12030269. [PMID: 35323743 PMCID: PMC8950343 DOI: 10.3390/membranes12030269] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023]
Abstract
Isobavachalcone (IBC) is a natural prenylated chalcone with a broad spectrum of pharmacological properties. In this work, we newly synthesized and investigated the antibacterial activity of IBC against Gram-positive, Gram-negative and mycobacterial species. IBC was active against Gram-positive bacteria, mainly against Methicillin-Susceptible Staphylococcus aureus (MSSA) and Methicillin-Resistant Staphylococcus aureus (MRSA), with minimum inhibitory concentration (MIC) values of 1.56 and 3.12 µg/mL, respectively. On the other hand, IBC was not able to act against Gram-negative species (MIC > 400 µg/mL). IBC displayed activity against mycobacterial species (MIC = 64 µg/mL), including Mycobacterium tuberculosis, Mycobacterium avium and Mycobacterium kansasii. IBC was able to inhibit more than 50% of MSSA and MRSA biofilm formation at 0.78 µg/mL. Its antibiofilm activity was similar to vancomycin, which was active at 0.74 µg/mL. In order to study the mechanism of the action by fluorescence microscopy, the propidium iodide (PI) and SYTO9 fluorophores indicated that IBC disrupted the membrane of Bacillus subtilis. Toxicity assays using human keratinocytes (HaCaT cell line) showed that IBC did not have the capacity to reduce the cell viability. These results suggested that IBC is a promising antibacterial agent with an elucidated mode of action and potential applications as an antibacterial drug and a medical device coating.
Collapse
|
181
|
de Aguiar FRM, Rocha LQ, Barboza MMO, Sampaio TL, Nogueira NAP, de Menezes RRPPB. Occurrence of methicillin-resistant Staphylococcus aureus (MRSA) in 'coalho' cheese produced in Brazil. J DAIRY RES 2022; 89:1-4. [PMID: 35184776 DOI: 10.1017/s0022029922000073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The experiments reported in this research communication analysed the presence of methicillin-resistant Staphylococcus aureus (MRSA) in 112 samples of 'coalho' cheese, from 56 dairy producing farms in 28 cities in all mesoregions of the State of Ceará, Brazil. To assess antimicrobial resistance we also examined the presence of genes encoding enterotoxins and toxic shock syndrome toxin, as well as the presence of the blaZ gene for β-lactamases, and resistance to oxacillin. The research found 69 isolates of S. aureus, of which 13.04% had the mecA gene encoding the penicillin-binding protein, which confers resistance to methicillin, in cheese samples from 6 different cities. This included the state capital, Fortaleza, which had the largest prevalence (23.19%) of mecA positive isolates. It was also found that 55.07% of the isolates of S. aureus had the blaZ gene, and 7.25% demonstrated resistance to oxacillin in the plate disc diffusion tests. We did not show the presence of isolates carrying toxigenic genes. The findings suggest that strict supervision of production processes in the dairy industry is necessary in all production scale processes, thus preventing contamination and possible problems for consumers.
Collapse
Affiliation(s)
- Felipe R M de Aguiar
- Department of Pharmaceutical Sciences, Federal University of Ceará, Fortaleza, Brazil
| | - Larissa Q Rocha
- Department of Pharmaceutical Sciences, Federal University of Ceará, Fortaleza, Brazil
| | | | - Tiago L Sampaio
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, Brazil
| | - Nádia A P Nogueira
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, Brazil
| | - Ramon R P P B de Menezes
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
182
|
Saber T, Samir M, El-Mekkawy RM, Ariny E, El-Sayed SR, Enan G, Abdelatif SH, Askora A, Merwad AMA, Tartor YH. Methicillin- and Vancomycin-Resistant Staphylococcus aureus From Humans and Ready-To-Eat Meat: Characterization of Antimicrobial Resistance and Biofilm Formation Ability. Front Microbiol 2022; 12:735494. [PMID: 35211098 PMCID: PMC8861318 DOI: 10.3389/fmicb.2021.735494] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Methicillin-resistant and vancomycin-resistant Staphylococcus aureus (MRSA and VRSA) are zoonotic life-threatening pathogens, and their presence in food raises a public health concern. Yet, scarce data are available regarding MRSA and VRSA in both ready-to-eat (RTE) meat and food handlers. This study was undertaken to determine the frequency, antimicrobial resistance, and biofilm-forming ability of MRSA and VRSA isolated from RTE meat (shawarma and burger) and humans (food handlers, and hospitalized patients) in Zagazig city, Sharkia Governorate, Egypt. We analyzed 176 samples (112 human samples: 72 from hospitalized patients and 40 from food handlers, 64 RTE meat samples: 38 from shawarma and 26 from burger). Using phenotypic, PCR-based identification of nuc gene and matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), 60 coagulase-positive S. aureus (COPS) isolates were identified in the samples as follow: RTE meat (15/64, 23.4%), hospitalized patients (33/72, 45.8%) and food handlers (12/40, 30%). All the COPS isolates were mecA positive (and thus were classified as MRSA) and multidrug resistant with multiple antibiotic resistance indices ranging from 0.25 to 0.92. Overall, resistance to cefepime (96.7%), penicillin (88.3%), were common, followed by ampicillin-sulbactam (65%), ciprofloxacin (55%), nitrofurontoin (51.7%), and gentamicin (43.3%). VRSA was detected in 30.3% of COPS hospitalized patient's isolates, 26.7% of COPS RTE meat isolates and 25% of COPS food handler's isolates. VanA, vanB, or both genes were detected in 64.7, 5.9, and 29.4% of all VAN-resistant isolates, respectively. The majority of the COPS isolates (50/60, 83.3%) have biofilm formation ability and harbored icaA (76%), icaD (74%), icaC (50%), and icaB (46%) biofilm-forming genes. The bap gene was not detected in any of the isolates. The ability of MRSA and VRSA isolates to produce biofilms in addition to being resistant to antimicrobials highlight the danger posed by these potentially virulent microorganisms persisting in RTE meat, food handlers, and patients. Taken together, good hygiene practices and antimicrobial surveillance plans should be strictly implemented along the food chain to reduce the risk of colonization and dissemination of MRSA and VRSA biofilm-producing strains.
Collapse
Affiliation(s)
- Taisir Saber
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Mohamed Samir
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Rasha M. El-Mekkawy
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Eman Ariny
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Sara Ramadan El-Sayed
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Gamal Enan
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Sawasn H. Abdelatif
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed Askora
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Abdallah M. A. Merwad
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Yasmine H. Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
183
|
Almeida L, Lopes N, Gaio V, Cavaleiro C, Salgueiro L, Silva V, Poeta P, Cerca N. Thymbra capitata
essential oil has a significant antimicrobial activity against methicillin‐resistant
Staphylococcus aureus
pre‐formed biofilms. Lett Appl Microbiol 2022; 74:787-795. [DOI: 10.1111/lam.13665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Luciana Almeida
- Centre of Biological Engineering (CEB) Laboratory of Research in Biofilms Rosário Oliveira (LIBRO) University of Minho Campus de Gualtar Braga Portugal
| | - Nathalie Lopes
- Centre of Biological Engineering (CEB) Laboratory of Research in Biofilms Rosário Oliveira (LIBRO) University of Minho Campus de Gualtar Braga Portugal
- LABBELS –Associate Laboratory Braga, Guimarães Portugal
| | - Vânia Gaio
- Centre of Biological Engineering (CEB) Laboratory of Research in Biofilms Rosário Oliveira (LIBRO) University of Minho Campus de Gualtar Braga Portugal
- LABBELS –Associate Laboratory Braga, Guimarães Portugal
| | - Carlos Cavaleiro
- Faculty of Pharmacy of the University of Coimbra University of Coimbra Coimbra Portugal
- CIEPQPF Department of Chemical Engineering Faculty of Sciences and Technology University of Coimbra Coimbra Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy of the University of Coimbra University of Coimbra Coimbra Portugal
- CIEPQPF Department of Chemical Engineering Faculty of Sciences and Technology University of Coimbra Coimbra Portugal
| | - Vanessa Silva
- Microbiology and Antibiotic Resistance Team (MicroART) Department of Veterinary Sciences University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Veterinary and Animal Research Centre Associate Laboratory for Animal and Veterinary Science (AL4AnimalS) University of Trás‐os‐Montes and Alto Douro (UTAD) 5000‐801 Vila Real Portugal
- Associated Laboratory for Green Chemistry (LAQV‐REQUIMTE) University NOVA of Lisboa Lisboa, Caparica Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART) Department of Veterinary Sciences University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Veterinary and Animal Research Centre Associate Laboratory for Animal and Veterinary Science (AL4AnimalS) University of Trás‐os‐Montes and Alto Douro (UTAD) 5000‐801 Vila Real Portugal
- Associated Laboratory for Green Chemistry (LAQV‐REQUIMTE) University NOVA of Lisboa Lisboa, Caparica Portugal
| | - Nuno Cerca
- Centre of Biological Engineering (CEB) Laboratory of Research in Biofilms Rosário Oliveira (LIBRO) University of Minho Campus de Gualtar Braga Portugal
- LABBELS –Associate Laboratory Braga, Guimarães Portugal
| |
Collapse
|
184
|
Xiang YZ, Wu G, Yang LY, Yang XJ, Zhang YM, Lin LB, Deng XY, Zhang QL. Antibacterial effect of bacteriocin XJS01 and its application as antibiofilm agents to treat multidrug-resistant Staphylococcus aureus infection. Int J Biol Macromol 2022; 196:13-22. [PMID: 34838856 DOI: 10.1016/j.ijbiomac.2021.11.136] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/09/2021] [Accepted: 11/20/2021] [Indexed: 12/31/2022]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus biofilms have emerged as a serious threat to human health. Recently, the development of antibiotic replacement therapy has gained much attention due to the potential application of bacteriocin. The present study sought to evaluate the antibacterial effect of bacteriocin XJS01 against MDR S. aureus, a previously reported bacteriocin against S. aureus strain 2612:1606BL1486 (S. aureus_26, an MDR strain demonstrated here), and its potential application as an antibiofilm agent. The minimum bactericide concentration of XJS01 against MDR S. aureus_26 was 33.18 μg/mL. XJS01 exhibited excellent storage stability and resistance against acid and reduced the density of established MDR S. aureus_26 biofilm. The hemolytic and HEK293T cytotoxicity activities of XJS01 and the histological analyses in mice confirmed its safety. Moreover, XJS01 effectively disrupted the MDR S. aureus_26 biofilm established on the skin wound surface and reduced the biofilm-isolated bacteria, thereby decreasing the release of pro-inflammatory cytokines and the proliferation of alternatively activated macrophages. Compared to mupirocin, XJS01 exhibited an excellent therapeutic effect on mice skin wounds, confirming it to be a potential alternative to antibiotics.
Collapse
Affiliation(s)
- Yi-Zhou Xiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Gang Wu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Department of Neurology, Yan'an Hospital of Kunming City, Kunming, Yunnan 650051, China
| | - Lin-Yu Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xiao-Jie Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yan-Mei Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Lian-Bing Lin
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xian-Yu Deng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Qi-Lin Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|
185
|
Nadar S, Khan T, Patching SG, Omri A. Development of Antibiofilm Therapeutics Strategies to Overcome Antimicrobial Drug Resistance. Microorganisms 2022; 10:microorganisms10020303. [PMID: 35208758 PMCID: PMC8879831 DOI: 10.3390/microorganisms10020303] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
A biofilm is a community of stable microorganisms encapsulated in an extracellular matrix produced by themselves. Many types of microorganisms that are found on living hosts or in the environment can form biofilms. These include pathogenic bacteria that can serve as a reservoir for persistent infections, and are culpable for leading to a broad spectrum of chronic illnesses and emergence of antibiotic resistance making them difficult to be treated. The absence of biofilm-targeting antibiotics in the drug discovery pipeline indicates an unmet opportunity for designing new biofilm inhibitors as antimicrobial agents using various strategies and targeting distinct stages of biofilm formation. The strategies available to control biofilm formation include targeting the enzymes and proteins specific to the microorganism and those involved in the adhesion pathways leading to formation of resistant biofilms. This review primarily focuses on the recent strategies and advances responsible for identifying a myriad of antibiofilm agents and their mechanism of biofilm inhibition, including extracellular polymeric substance synthesis inhibitors, adhesion inhibitors, quorum sensing inhibitors, efflux pump inhibitors, and cyclic diguanylate inhibitors. Furthermore, we present the structure–activity relationships (SAR) of these agents, including recently discovered biofilm inhibitors, nature-derived bioactive scaffolds, synthetic small molecules, antimicrobial peptides, bioactive compounds isolated from fungi, non-proteinogenic amino acids and antibiotics. We hope to fuel interest and focus research efforts on the development of agents targeting the uniquely complex, physical and chemical heterogeneous biofilms through a multipronged approach and combinatorial therapeutics for a more effective control and management of biofilms across diseases.
Collapse
Affiliation(s)
- Sahaya Nadar
- Department of Pharmaceutical Chemistry, St. John Institute of Pharmacy and Research, Mumbai 400056, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India;
| | - Simon G. Patching
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: or (S.G.P.); (A.O.)
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: or (S.G.P.); (A.O.)
| |
Collapse
|
186
|
New update on molecular diversity of clinical Staphylococcus aureus isolates in Iran: antimicrobial resistance, adhesion and virulence factors, biofilm formation and SCCmec typing. Mol Biol Rep 2022; 49:3099-3111. [PMID: 35064407 DOI: 10.1007/s11033-022-07140-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/10/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Staphylococcus aureus is often considered as a potential pathogen and resistant to a wide range of antibiotics. The pathogenicity of this bacterium is due to the presence of multiple virulence factors and the ability to form biofilm. SCCmec types I, II and III are mainly attributed to HA-MRSA, while SCCmec types IV and V have usually been reported in CA-MRSA infections. METHODS AND RESULTS In this study, we performed a cross-sectional study to determine the antimicrobial resistance, adhesion and virulence factors, biofilm formation and SCCmec typing of clinical S. aureus isolates in Iran. S. aureus isolates were identified using microbiological standard methods and antibiotic susceptibility tests were performed as described by the Clinical and Laboratory Standards Institute (CLSI) guidelines. Inducible resistance phenotype and biofilm formation were determined using D-test and tissue culture plate methods, respectively. Multiplex-PCRs were performed to detect adhesion and virulence factors, antibiotic resistance genes, biofilm formation and SCCmec typing by specific primers. Among 143 clinical samples, 67.8% were identified as MRSA. All isolates were susceptible to vancomycin. The prevalence of cMLSB, iMLSB and MS phenotypes were 61.1%, 22.2% and 14.8%, respectively. The TCP method revealed that 71.3% of isolates were able to form biofilm. The predominant virulence and inducible resistance genes in both MRSA and MSSA isolates were related to sea and ermC respectively. SCCmec type III was the predominant type. CONCLUSIONS Data show the high prevalence rates of virulence elements among S. aureus isolates, especially MRSA strains. This result might be attributed to antibiotic pressure, facilitating clonal selection.
Collapse
|
187
|
Matsumoto M, Kozakai M, Furuta RA, Matsubayashi K, Satake M. Association of Staphylococcus aureus in platelet concentrates with skin diseases in blood donors: Limitations of cultural bacterial screening. Transfusion 2022; 62:621-632. [PMID: 35045189 DOI: 10.1111/trf.16804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Bacterial contamination in platelet concentrates (PCs) is a major problem in transfusion medicine. Contamination with Staphylococcus aureus is occasionally missed, even with cultural screening. STUDY DESIGN AND METHODS Donors implicated in S. aureus-contaminated PC were followed up. Skin and nasal swab specimens from six donors and S. aureus isolated from PCs related to these donors were subjected to multilocus sequence typing (MLST) and pulsed-field gel electrophoresis (PFGE) to determine the identity of bacteria. To evaluate the validity of the screening method using BacT/ALERT 3D, we spiked S. aureus and three other bacterial species as comparisons into PCs and investigated their growth pattern. RESULTS S. aureus was isolated from all nasal specimens and from the arm skin specimens of three donors with atopic dermatitis. In all cases, the S. aureus strains isolated from the PC and those from the nasal and skin specimens of the same donor showed concordant results using MLST and PFGE. In the spiking study, S. aureus showed irregular detectability over 24 to 48 h post-spike periods, whereas the three other bacterial species were detected in all culture bottles after a 24-h post-spike period. DISCUSSION The strain identity of S. aureus between donor and PC suggests that the contaminants were derived from those colonized in the donor. Furthermore, S. aureus yielded false-negative results using BacT/ALERT 3D.
Collapse
Affiliation(s)
- Mami Matsumoto
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Moe Kozakai
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Rika A Furuta
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Keiji Matsubayashi
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Masahiro Satake
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| |
Collapse
|
188
|
Bouchelaghem S, Das S, Naorem RS, Czuni L, Papp G, Kocsis M. Evaluation of Total Phenolic and Flavonoid Contents, Antibacterial and Antibiofilm Activities of Hungarian Propolis Ethanolic Extract against Staphylococcus aureus. Molecules 2022; 27:574. [PMID: 35056886 PMCID: PMC8782033 DOI: 10.3390/molecules27020574] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022] Open
Abstract
Propolis is a natural bee product that is widely used in folk medicine. This study aimed to evaluate the antimicrobial and antibiofilm activities of ethanolic extract of propolis (EEP) on methicillin-resistant and sensitive Staphylococcus aureus (MRSA and MSSA). Propolis samples were collected from six regions in Hungary. The minimum inhibitory concentrations (MIC) values and the interaction of EEP-antibiotics were evaluated by the broth microdilution and the chequerboard broth microdilution methods, respectively. The effect of EEP on biofilm formation and eradication was estimated by crystal violet assay. Resazurin/propidium iodide dyes were applied for simultaneous quantification of cellular metabolic activities and dead cells in mature biofilms. The EEP1 sample showed the highest phenolic and flavonoid contents. The EEP1 successfully prevented the growth of planktonic cells of S. aureus (MIC value = 50 µg/mL). Synergistic interactions were shown after the co-exposition to EEP1 and vancomycin at 108 CFU/mL. The EEP1 effectively inhibited the biofilm formation and caused significant degradation of mature biofilms (50-200 µg/mL), as a consequence of the considerable decrement of metabolic activity. The EEP acts effectively as an antimicrobial and antibiofilm agent on S. aureus. Moreover, the simultaneous application of EEP and vancomycin could enhance their effect against MRSA infection.
Collapse
Affiliation(s)
- Sarra Bouchelaghem
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Sourav Das
- Department of Laboratory Medicine, Medical School, University of Pécs, Ifjúság Str. 13, 7624 Pécs, Hungary;
| | - Romen Singh Naorem
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Lilla Czuni
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Gábor Papp
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Marianna Kocsis
- Department of Plant Biology, Institute of Biology, University of Pécs, Ifjúság str. 6, 7624 Pécs, Hungary
| |
Collapse
|
189
|
Cates NK, Bunka TJ, Kavanagh AM, Wynes J. Split Anterior Tibial Tendon Transfer to Dorsal Lateral Foot for Cavovarus Deformities With Neuropathic Ulcerations: A Case Series. J Foot Ankle Surg 2022; 61:189-194. [PMID: 34489166 DOI: 10.1053/j.jfas.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/03/2023]
Abstract
Cavovarus deformity leads to increased peak pressure on the plantar lateral foot, which can lead to ulceration, and can potentially progress to amputation. Techniques have been suggested in the treatment of cavovarus deformity, such as peroneus brevis or longus tendon transfer, anterior tibial tendon lengthening, posterior tibial tendon transfer, or boney resection. This case series shows split anterior tibial tendon transfer as a surgical reconstruction of cavovarus pedal deformity. Our technique of split anterior tibial tendon in-phase transfer to the dorsal lateral foot, restores the eversion and dorsiflexory pull necessary to offset peroneal attenuation. The procedure can be performed primarily or staged, in order to achieve infection temporization prior to the transfer. A total of 14 patients underwent split anterior tibial tendon transfer, 57.14% (8/14) of which had preoperative ulcerations, and 42.86% (6/14) of which had preoperative hyperkeratotic pre-ulcerative lesions. The preoperative ulcerations were present for an average of 67.89 weeks (range 2-232), with an average area of 6.09 ± 7.44 cm2. The ulcerations healed in 75% (6/8) of the patients, at 19.67 weeks (range 1.57-76), with new ulceration occurrence in 7.14% (1/14) of patients, 7.14% (1/14) rate of ulceration recurrence. None of the patients went on to minor or major amputation. The goal of the tendon transfer is to decrease midfoot plantar pressures on the lateral foot and allow for resolution of pre-existing ulcerations and rebalancing the foot and ankle.
Collapse
Affiliation(s)
- Nicole K Cates
- Fellowship Trained Foot and Ankle Surgeon, Hand & Microsurgery Medical Group, San Francisco, CA.
| | - Taylor J Bunka
- Resident Physician, Department of Plastics Surgery, MedStar Georgetown University Hospital, Washington, DC
| | - Amber M Kavanagh
- Resident Physician, Department of Plastics Surgery, MedStar Georgetown University Hospital, Washington, DC
| | - Jacob Wynes
- Assistant Professor Fellowship Program Director, Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
190
|
da Cruz Nizer WS, Ferraz AC, Moraes TDFS, Lima WG, Dos Santos JP, Duarte LP, Magalhães CLDB, Vieira-Filho SA, de Magalhães JC. Netzahualcoyonol from Salacia multiflora (Lam.) DC. (Celastraceae) roots as a bioactive compound against gram-positive pathogens. Nat Prod Res 2022; 36:5904-5909. [PMID: 34994265 DOI: 10.1080/14786419.2021.2023865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The expression of virulence factors, such as biofilm formation, in association with the acquisition of resistance to multiple drugs, has evidenced the need for new and effective antimicrobial agents against Staphylococcus aureus. The evaluation of the pharmacological properties of plant-derived compounds is a promising alternative to the development of new antimicrobials. In this study, we aimed to evaluate the antibacterial, antibiofilm, and the synergistic and cytotoxic effects of netzahualcoyonol isolated from Salacia multiflora (Lam.) DC. roots. Netzahualcoyonol presented bacteriostatic (1.56-25.0 µg/mL) and bactericidal (25.0-400.0 µg/mL) effects against Gram-positive bacteria, disrupted the biofilm of S. aureus, and presented a synergistic effect after its combination with β-lactams and aminoglycosides. The low cytotoxicity of netzahualcoyonol (Selectivity Index (SI) for S. aureus (2.56), S. saprophyticus (20.56), and Bacillus subtilis (1.28)) suggests a good security profile. Taken together, these results show that netzahualcoyonol is promising for the development of a new effective antibacterial agent.
Collapse
Affiliation(s)
- Waleska Stephanie da Cruz Nizer
- Department of Chemistry, Biotechnology, and Bioprocess Engineering, Universidade Federal de São João del-Rei, Ouro Branco, MG, Brazil.,Carleton University, Ottawa, ON, Canada
| | - Ariane Coelho Ferraz
- Institute of Exact and Biological Sciences, Biological Science Research Nucleus, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | | | - William Gustavo Lima
- Laboratory of Medical Microbiology, Campus Centro Oeste Dona Lindu, Universidade Federal de São João del-Rei, Divinópolis, MG, Brazil
| | | | - Lucienir Pains Duarte
- Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cintia Lopes de Brito Magalhães
- Institute of Exact and Biological Sciences, Biological Science Research Nucleus, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | | | - José Carlos de Magalhães
- Department of Chemistry, Biotechnology, and Bioprocess Engineering, Universidade Federal de São João del-Rei, Ouro Branco, MG, Brazil
| |
Collapse
|
191
|
Kurosu M, Mitachi K, Yang J, Pershing EV, Horowitz BD, Wachter EA, Lacey JW, Ji Y, Rodrigues DJ. Antibacterial Activity of Pharmaceutical-Grade Rose Bengal: An Application of a Synthetic Dye in Antibacterial Therapies. Molecules 2022; 27:322. [PMID: 35011554 PMCID: PMC8746496 DOI: 10.3390/molecules27010322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 11/27/2022] Open
Abstract
Rose bengal has been used in the diagnosis of ophthalmic disorders and liver function, and has been studied for the treatment of solid tumor cancers. To date, the antibacterial activity of rose bengal has been sporadically reported; however, these data have been generated with a commercial grade of rose bengal, which contains major uncontrolled impurities generated by the manufacturing process (80-95% dye content). A high-purity form of rose bengal formulation (HP-RBf, >99.5% dye content) kills a battery of Gram-positive bacteria, including drug-resistant strains at low concentrations (0.01-3.13 μg/mL) under fluorescent, LED, and natural light in a few minutes. Significantly, HP-RBf effectively eradicates Gram-positive bacterial biofilms. The frequency that Gram-positive bacteria spontaneously developed resistance to HP-RB is extremely low (less than 1 × 10-13). Toxicity data obtained through our research programs indicate that HP-RB is feasible as an anti-infective drug for the treatment of skin and soft tissue infections (SSTIs) involving multidrug-resistant (MDR) microbial invasion of the skin, and for eradicating biofilms. This article summarizes the antibacterial activity of pharmaceutical-grade rose bengal, HP-RB, against Gram-positive bacteria, its cytotoxicity against skin cells under illumination conditions, and mechanistic insights into rose bengal's bactericidal activity under dark conditions.
Collapse
Affiliation(s)
- Michio Kurosu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA;
| | - Katsuhiko Mitachi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA;
| | - Junshu Yang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 205 VSB, 1971 Commonwealth Avenue, Saint Paul, MN 55108, USA; (J.Y.); (Y.J.)
| | - Edward V. Pershing
- Provectus Biopharmaceuticals, Inc., 10025 Investment Drive, Suite 250, Knoxville, TN 37932, USA; (E.V.P.); (B.D.H.); (E.A.W.); (J.W.L.III); (D.J.R.)
| | - Bruce D. Horowitz
- Provectus Biopharmaceuticals, Inc., 10025 Investment Drive, Suite 250, Knoxville, TN 37932, USA; (E.V.P.); (B.D.H.); (E.A.W.); (J.W.L.III); (D.J.R.)
| | - Eric A. Wachter
- Provectus Biopharmaceuticals, Inc., 10025 Investment Drive, Suite 250, Knoxville, TN 37932, USA; (E.V.P.); (B.D.H.); (E.A.W.); (J.W.L.III); (D.J.R.)
| | - John W. Lacey
- Provectus Biopharmaceuticals, Inc., 10025 Investment Drive, Suite 250, Knoxville, TN 37932, USA; (E.V.P.); (B.D.H.); (E.A.W.); (J.W.L.III); (D.J.R.)
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 205 VSB, 1971 Commonwealth Avenue, Saint Paul, MN 55108, USA; (J.Y.); (Y.J.)
| | - Dominic J. Rodrigues
- Provectus Biopharmaceuticals, Inc., 10025 Investment Drive, Suite 250, Knoxville, TN 37932, USA; (E.V.P.); (B.D.H.); (E.A.W.); (J.W.L.III); (D.J.R.)
| |
Collapse
|
192
|
Li Q, Yu S, Han J, Wu J, You L, Shi X, Wang S. Synergistic antibacterial activity and mechanism of action of nisin/carvacrol combination against Staphylococcus aureus and their application in the infecting pasteurized milk. Food Chem 2022; 380:132009. [PMID: 35077986 DOI: 10.1016/j.foodchem.2021.132009] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/14/2021] [Accepted: 12/14/2021] [Indexed: 11/04/2022]
Abstract
Synergistic antibacterial effect is a promising way to overcome the challenge of microbial contamination in food. In this study, we detected the synergistic interactions of nisin and carvacrol. The MIC of nisin and carvacrol against S. aureus were 60 and 125 μg/mL, respectively. The FICI and FBCI were 0.28125 and 0.09375, which suggested that the nisin/carvacrol combination presented synergistic antibacterial effect against S. aureus. The antibacterial activity of nisin/carvacrol combination was much higher than their individuals and the dose of antibacterials was obviously reduced. The combination could completely kill S. aureus within 8 h, accelerate the destruction of cell membrane, and inhibit formation of biofilm. Under the intervention of nisin, more CAR could enter cell to hunt intracellular targets, leading to an increase in intracellular antibacterial level. Besides, in the storage of pasteurized milk, the combinational treatment successfully inhibited microbial reproduction at 25 °C and 4 °C. Thus, the combination of nisin and carvacrol was a potential synergistic strategy for food preservation.
Collapse
Affiliation(s)
- Qingxiang Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China
| | - Shuna Yu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China
| | - Jinzhi Han
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China
| | - Jiulin Wu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China.
| | - Lijun You
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China
| | - Xiaodan Shi
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China
| | - Shaoyun Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, PR China.
| |
Collapse
|
193
|
Yang M, Zhang J, Wei Y, Zhang J, Tao C. Recent advances in metal-organic framework-based materials for anti-staphylococcus aureus infection. NANO RESEARCH 2022; 15:6220-6242. [PMID: 35578616 PMCID: PMC9094125 DOI: 10.1007/s12274-022-4302-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 05/03/2023]
Abstract
The rapid spread of staphylococcus aureus (S. aureus) causes an increased morbidity and mortality, as well as great economic losses in the world. Anti-S. aureus infection becomes a major challenge for clinicians and nursing professionals to address drug resistance. Hence, it is urgent to explore high efficiency, low toxicity, and environmental-friendly methods against S. aureus. Metal-organic frameworks (MOFs) represent great potential in treating S. aureus infection due to the unique features of MOFs including tunable chemical constitute, open crystalline structure, and high specific surface area. Especially, these properties endow MOF-based materials outstanding antibacterial effect, which can be mainly attributed to the continuously released active components and the exerted catalytic activity to fight bacterial infection. Herein, the structural characteristics of MOFs and evaluation method of antimicrobial activity are briefly summarized. Then we systematically give an overview on their recent progress on antibacterial mechanisms, metal ion sustained-release system, controlled delivery system, catalytic system, and energy conversion system based on MOF materials. Finally, suggestions and direction for future research to develop and mechanism understand MOF-based materials are discussed in antibacterial application.
Collapse
Affiliation(s)
- Mei Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Jin Zhang
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065 China
| | - Yinhao Wei
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Jie Zhang
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065 China
| | - Chuanmin Tao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041 China
| |
Collapse
|
194
|
Risks and Prevention of Surgical Site Infection After Hernia Mesh Repair and the Predictive Utility of ACS-NSQIP. J Gastrointest Surg 2022; 26:950-964. [PMID: 35064459 PMCID: PMC9021144 DOI: 10.1007/s11605-022-05248-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023]
Abstract
AIM The aim of this paper was to provide a narrative review of surgical site infection after hernia surgery and the influence of perioperative preventative interventions. METHODS The review was based on current national and international guidelines and a literature search. RESULTS Mesh infection is a highly morbid complication after hernia surgery, and is associated with hospital re-admission, increased health care costs, re-operation, hernia recurrence, impaired quality of life and plaintiff litigation. The American College of Surgeons National Surgical Quality Improvement Program is a particularly useful resource for the study and evidence-based practise of abdominal wall hernia repair. DISCUSSION The three major modifiable patient comorbidities significantly associated with postoperative surgical site infection in hernia surgery are obesity, tobacco smoking and diabetes mellitus. Preoperative optimization includes weight loss, cessation of smoking, and control of diabetes. Intraoperative interventions relate, in particular, to the control of fomite mediated transmission in the operating theatre and prevention of mesh contamination with S. aureus CFUs. Risk management strategies should also target the niche ecological conditions which enable bacterial survival and subsequent biofilm formation on an implanted mesh. Outcomes of mesh infection after hernia surgery are closely related to mesh type and porosity, patient smoking status, presence of MRSA, bacterial adhesion and biofilm production. The use of suction drains and the timing of drain removal are controversial and discussed in detail. Finally, the utility of the ACS-NSQIP Surgical Risk Calculator in predicting complications and outcomes in individual patients and the importance of quality improvement initiatives in surgical units are emphasized.
Collapse
|
195
|
Carrera-Salinas A, González-Díaz A, Vázquez-Sánchez DA, Camoez M, Niubó J, Càmara J, Ardanuy C, Martí S, Domínguez MÁ, Garcia M, Marco F, Chaves F, Cercenado E, Tapiol J, Xercavins M, Fontanals D, Loza E, Rodríguez-López F, Olarte I, Mirelis B, Ruiz de Gopegui E, Lepe J, Larrosa N. Staphylococcus aureus surface protein G (sasG) allelic variants: correlation between biofilm formation and their prevalence in methicillin-resistant S. aureus (MRSA) clones. Res Microbiol 2022; 173:103921. [DOI: 10.1016/j.resmic.2022.103921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/02/2023]
|
196
|
Hong W, Nyaruaba R, Li X, Liu H, Yang H, Wei H. In-situ and Real-Time Monitoring of the Interaction Between Lysins and Staphylococcus aureus Biofilm by Surface Plasmon Resonance. Front Microbiol 2021; 12:783472. [PMID: 34917062 PMCID: PMC8670000 DOI: 10.3389/fmicb.2021.783472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022] Open
Abstract
Staphylococcus aureus can produce a multilayered biofilm embedded in extracellular polymeric matrix. This biofilm is difficult to remove, insensitive to antibiotics, easy to develop drug-resistant strains and causes enormous problems to environments and health. Phage lysin which commonly consists of a catalytic domain (CD) and a cell-wall binding domain (CBD) is a powerful weapon against bacterial biofilm. However, the real-time interaction between lysin and S. aureus biofilm is still not fully understood. In this study, we monitored the interactions of three lysins (ClyF, ClyC, PlySs2) against culture-on-chip S. aureus biofilm, in real-time, based on surface plasmon resonance (SPR). A typical SPR response curve showed that the lysins bound to the biofilm rapidly and the biofilm destruction started at a longer time. By using 1:1 binding model analysis, affinity constants (KD) for ClyF, ClyC, and PlySs2 were found to be 3.18 ± 0.127 μM, 1.12 ± 0.026 μM, and 15.5 ± 0.514 μM, respectively. The fact that ClyF and PlySs2 shared the same CBD but showed different affinity to S. aureus biofilm suggested that, not only CBD, but also CD affects the binding activity of the entire lysin. The SPR platform can be applied to improve our understanding on the complex interactions between lysins and bacterial biofilm including association (adsorption) and disassociation (destruction).
Collapse
Affiliation(s)
- Wei Hong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Raphael Nyaruaba
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaohong Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Huan Liu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Yang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hongping Wei
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
197
|
Nguyen TK, Peyrusson F, Siala W, Pham NH, Nguyen HA, Tulkens PM, Van Bambeke F. Activity of Moxifloxacin Against Biofilms Formed by Clinical Isolates of Staphylococcus aureus Differing by Their Resistant or Persister Character to Fluoroquinolones. Front Microbiol 2021; 12:785573. [PMID: 34975808 PMCID: PMC8715871 DOI: 10.3389/fmicb.2021.785573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus biofilms are poorly responsive to antibiotics. Underlying reasons include a matrix effect preventing drug access to embedded bacteria, or the presence of dormant bacteria with reduced growth rate. Using 18 clinical isolates previously characterized for their moxifloxacin-resistant and moxifloxacin-persister character in stationary-phase culture, we studied their biofilm production and matrix composition and the anti-biofilm activity of moxifloxacin. Biofilms were grown in microtiter plates and their abundance quantified by crystal violet staining and colony counting; their content in polysaccharides, extracellular DNA and proteins was measured. Moxifloxacin activity was assessed after 24 h of incubation with a broad range of concentrations to establish full concentration-response curves. All clinical isolates produced more biofilm biomass than the reference strain ATCC 25923, the difference being more important for those with high relative persister fractions to moxifloxacin, most of which being also resistant. High biofilm producers expressed icaA to higher levels, enriching the matrix in polysaccharides. Moxifloxacin was less potent against biofilms from clinical isolates than from ATCC 25923, especially against moxifloxacin-resistant isolates with high persister fractions, which was ascribed to a lower concentration of moxifloxacin in these biofilms. Time-kill curves in biofilms revealed the presence of a moxifloxacin-tolerant subpopulation, with low multiplication capacity, whatever the persister character of the isolate. Thus, moxifloxacin activity depends on its local concentration in biofilm, which is reduced in most isolates with high-relative persister fractions due to matrix effects, and insufficient to kill resistant isolates due to their high MIC.
Collapse
Affiliation(s)
- Tiep K. Nguyen
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Department of Pharmaceutical Industry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Frédéric Peyrusson
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Wafi Siala
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nhung H. Pham
- Department of Microbiology, Bach Mai Hospital, Hanoi, Vietnam
| | - Hoang A. Nguyen
- The National Center for Drug Information and Adverse Drug Reactions Monitoring, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Paul M. Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Françoise Van Bambeke,
| |
Collapse
|
198
|
Arenas J, Szabo Z, van der Wal J, Maas C, Riaz T, Tønjum T, Tommassen J. Serum proteases prevent bacterial biofilm formation: role of kallikrein and plasmin. Virulence 2021; 12:2902-2917. [PMID: 34903146 PMCID: PMC8677018 DOI: 10.1080/21505594.2021.2003115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Biofilm formation is a general strategy for bacterial pathogens to withstand host defense mechanisms. In this study, we found that serum proteases inhibit biofilm formation by Neisseria meningitidis, Neisseria gonorrhoeae, Haemophilus influenzae, and Bordetella pertussis. Confocal laser-scanning microscopy analysis revealed that these proteins reduce the biomass and alter the architecture of meningococcal biofilms. To understand the underlying mechanism, the serum was fractionated through size-exclusion chromatography and anion-exchange chromatography, and the composition of the fractions that retained anti-biofilm activity against N. meningitidis was analyzed by intensity-based absolute quantification mass spectrometry. Among the identified serum proteins, plasma kallikrein (PKLK), FXIIa, and plasmin were found to cleave neisserial heparin-binding antigen and the α-peptide of IgA protease on the meningococcal cell surface, resulting in the release of positively charged polypeptides implicated in biofilm formation by binding extracellular DNA. Further experiments also revealed that plasmin and PKLK inhibited biofilm formation of B. pertussis by cleaving filamentous hemagglutinin. We conclude that the proteolytic activity of serum proteases toward bacterial adhesins involved in biofilm formation could constitute a defense mechanism for the clearance of pathogens.
Collapse
Affiliation(s)
- Jesús Arenas
- Department of Molecular Microbiology and Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands.,Unit of Microbiology and Immunology, Faculty of Veterinary, University of Zaragoza, Zaragoza, Spain
| | - Zalan Szabo
- Research and Development Department, U-Protein Express BV, Utrecht, The Netherlands
| | - Jelle van der Wal
- Department of Molecular Microbiology and Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands
| | - Coen Maas
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tahira Riaz
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Tone Tønjum
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Jan Tommassen
- Department of Molecular Microbiology and Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
199
|
Zhu M, Wang ZJ, He YJ, Qin Y, Zhou Y, Qi ZH, Zhou ZS, Zhu YY, Jin DN, Chen SS, Luo XD. Bioguided isolation, identification and bioactivity evaluation of anti-MRSA constituents from Morus alba Linn. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114542. [PMID: 34428525 DOI: 10.1016/j.jep.2021.114542] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE The root bark of Morus alba Linn. (M. alba), a traditional folk medicine, has been documented in the Chinese Pharmacopoeia, which has been widely used for asthma, fever, pneumonia, edema, vomit, colitis, bronchitis and keratitis diseases. Some of the diseases may be related to respiratory, digestive, urinary tract infections. Although Diels-Alder adducts (DAAs), flavonoids, 2-arylbenzofurans and stilbene compounds have been isolated from the root bark of M. alba, few compounds are reported for their antimicrobial efficacy in vivo and the mechanism. AIM OF THE STUDY The aim of the study was to isolate and identify compounds of the root bark of M. alba in view of their anti-MRSA bioactivity, evaluate the anti-MRSA bioactivity of compounds and 60% ethanol elution (MA-6) in vitro and in vivo, and explore preliminary antibacterial mechanism in order to provide natural resources against MRSA infection. MATERIALS AND METHODS Systematic phytochemical investigations were carried out according to the thin layer chromatography (TLC) of the active fraction MA-6 to find more anti-MRSA ingredients. The compounds of the root bark of M. alba were separated by column chromatography and identified by LC-MS/MS and NMR spectroscopy. The anti-MRSA efficacy of the active ingredients were evaluated by broth microdilution method and a murine infection model. The mode of action of compounds was explored by time-kill curve and post-contact effect. The preliminary mechanism of compounds against MRSA was explored by drug efflux pumps and bacterial biofilms. RESULTS Chemical isolation resulted in twenty-nine known compounds, most with one or more geranyl and prenyl units exhibited superior anti-MRSA bioactivity, with MIC values of 2-16 μg/mL. In addition, the mode of action indicated that compounds presented persistent antimicrobial effect, which also produced concentration-dependent and time-dependent killing activity or property. Preliminary mechanism showed that the compound kuwanon O (29) damaged the bacterial cell membranes, leading to the accumulation of antibiotics inside bacterial cells, moreover, MA-6 and kuwanon O (29) inhibited the efflux of drugs by combining with methicillin or ethidium bromide (EtBr), resulting in the MICs of EtBr and methicillin were obviously decreased three-fold. The anti-MRSA efficacy in vivo indicated that the active fraction MA-6 could reduce bacteria in spleen, liver, kidney and mortality of acutely infectious mice, which was better than the positive drug berberine chloride. CONCLUSION Experimental investigation showed that the MA-6 and compound 29 have promising bioactivity against MRSA in vitro and in vivo, which might be used as a potential source of new antibacterial medicine or a potential efflux pump inhibitor against MRSA infection.
Collapse
Affiliation(s)
- Meng Zhu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Zhao-Jie Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Ying-Jie He
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Yan Qin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Ying Zhou
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Zi-Heng Qi
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Zhong-Shun Zhou
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Yan-Yan Zhu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Dan-Ni Jin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Shan-Shan Chen
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Xiao-Dong Luo
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China.
| |
Collapse
|
200
|
Bencardino D, Amagliani G, Brandi G. Carriage of Staphylococcus aureus among food handlers: An ongoing challenge in public health. Food Control 2021. [DOI: 10.1016/j.foodcont.2021.108362] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|