1
|
Li T, Shen H, Wang P, Wang Y, Liu J, He M, Gu L, Wang S, Wei M. Antibiofilm activity of arachidonic acid against linezolid-resistant Enterococcus faecalis: A potential strategy for combating biofilm-related infections. Microb Pathog 2025; 205:107629. [PMID: 40287107 DOI: 10.1016/j.micpath.2025.107629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/12/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Enterococcus faecalis, a prevalent opportunistic pathogen, readily forms biofilms on surfaces, contributing to its virulence and antibiotic resistance. Previous studies have highlighted arachidonic acid (AA) as a promising antibiofilm agent. This investigation aimed to elucidate the antibiofilm activity of AA against linezolid-resistant E. faecalis and explore its potential molecular mechanisms. A total of 21 E. faecalis strains were included in this study: the standard strain ATCC 29212 and 20 linezolid-resistant clinical isolates. To determine the minimum inhibitory concentrations (MICs) of antibiotics and AA, the microbroth dilution method was performed. Checkerboard microdilution assays were also performed to assess the interaction between AA and linezolid. The crystal violet method was performed to evaluate the biofilm formation abilities of E. faecalis and the biofilm inhibition activities of different concentrations of AA (0.25 mM, 0.5 mM, and 1 mM). In addition, the expression levels of biofilm-related genes (ebpA, ebpB, ebpC, gelE, ace, atlA, and esp) were measured employing quantitative reverse transcription PCR (qRT-PCR). All clinical strains exhibited resistance to linezolid and tetracycline while remaining susceptible to other antibiotics, including penicillin, ampicillin, vancomycin, teicoplanin, and tigecycline. The MICs of AA against all 21 E. faecalis isolates were >1 mM. Synergy was observed in 75 % of strains (15/20), while additivity was observed in 25 % strains (5/20). All isolates displayed strong biofilm-forming abilities. AA significantly reduced biofilm formation with average inhibition rates of 68.16 %, 69.64 %, and 72.01 % at concentrations of 0.25 mM, 0.5 mM, and 1 mM, respectively (P < 0.001). AA treatment resulted in decreased expression of ebpB, ebpC, ace, and atlA (P < 0.001), while simultaneously increasing gelE expression (P < 0.05). In conclusion, AA exhibited potent inhibitory effects on E. faecalis biofilm formation probably by influencing bacterial adhesion. The addition of AA can restore the susceptibility of linezolid against E. faecalis. These findings suggest that AA may serve as a potential therapeutic agent for preventing and treating E. faecalis infections, particularly those associated with biofilm formation.
Collapse
Affiliation(s)
- Tianmeng Li
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China
| | - Hong Shen
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China
| | - Peng Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China
| | - Yu Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China
| | - Jun Liu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China
| | - Meibo He
- Peking University Medical Press, China
| | - Li Gu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China.
| | - Shuai Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China.
| | - Ming Wei
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, China.
| |
Collapse
|
2
|
Senthil Kumar SA, Praveenkumar K, Jothipandian S, Swaroop S, Nithyanand P. Nanoscale surface modifications on Titanium plates- A strategy to mitigate MRSA biofilm-mediated implant infections: A pilot study. Microb Pathog 2025; 203:107481. [PMID: 40089195 DOI: 10.1016/j.micpath.2025.107481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Orthopaedic implant infections pose a major threat after implantation. Biofilms of pathogenic bacteria resistant to antibiotics cause biomaterial mediated infections. Methicillin-resistant Staphylococcus aureus (MRSA) is one of the prevalent biofilm-forming pathogens associated with implant infection in high proportion. Loss of effectiveness of antibiotics against these drug-resistant pathogens demands alternative approaches to surmount this crisis. Various strategies involving antibiotics, biocides, and metal ions are employed as the prohibiting steps of biofilm formation. Hence, to prevent biofilm formation and infections caused by biofilms formed over the orthopaedic implants, we involved laser micro-machining to modify the surface of the Titanium (Ti) plate, the most widely used implant material. Interestingly, we found that the laser-peening process generated widespread nanosized pores and micro-roughness to the surface of the Ti plate. Laser-peened Ti plate reduced the adhesion of MRSA over the metal surface and also retained its capacity to inhibit biofilm formation, which was confirmed with scanning electron microscopy (SEM). The biofilm assays like quantification of biofilm by crystal violet, determination of colony forming unit from biofilm formed over the control and laser-peened Ti plates showed that the laser-peened Ti plate significantly reduced the adherence of biofilm-forming MRSA. Moreover, the genes responsible for biofilm adhesion were found to be downregulated which was confirmed by qPCR. From our results, it was found that laser-peened Ti implants would be an alternative strategy to prevent biofilm-mediated infection on orthopaedic implant material.
Collapse
Affiliation(s)
- Sudaarsan Aruna Senthil Kumar
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India
| | - K Praveenkumar
- Department of Materials Engineering, Indian Institute of Science, Banglore, 560012, India
| | - Sowndarya Jothipandian
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India
| | - S Swaroop
- Surface Modification Laboratory, School of Advanced Sciences, Vellore Institute of Technology, Vellore-632014, India.
| | - Paramasivam Nithyanand
- Biofilm Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India.
| |
Collapse
|
3
|
Hanafy MS, Kamal NM, Fathallah HA. Antibacterial efficacy of ultrasonically activated probiotic endodontic irrigant against Enterococcus faecalis biofilm: an in-vitro study. BMC Oral Health 2025; 25:794. [PMID: 40414833 DOI: 10.1186/s12903-025-06212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Enterococcus faecalis (E. faecalis) is the most frequently retrieved microorganism from teeth with failed endodontic treatment. Sodium hypochlorite (NaOCl) irrigant still poses some drawbacks, such as its cytotoxic effect and reduced effectiveness when applied at lower concentrations. Root canal disinfection by probiotics may yield positive outcomes due to their proven antibacterial and anti-inflammatory abilities. This research was intended to assess the antibacterial efficacy of a probiotic irrigant after ultrasonic activation against E. faecalis in a tooth model. METHODS Teeth specimens were infected with E. faecalis biofilm and then randomly divided into five groups according to the final flush irrigation protocol used; PRO for probiotic irrigant, PRO + for activated probiotic irrigant, NaOCl for NaOCl irrigant, NaOCl + for activated NaOCl irrigant, and saline for saline irrigation. Activation of the irrigant was done for 1 min using an Ultra X ultrasonic tip. By counting the colony-forming units per milliliter, the antibacterial activity was quantitatively evaluated for each group pre- and post-irrigation application; then, the bacterial load reduction percentages were calculated accordingly. The one-way ANOVA was conducted to compare the mean values of all variables, followed by the post-hoc Tukey test to make group comparisons with a significance level set at p < 0.05. RESULTS All experimental groups exerted antibacterial activity against E. faecalis with a reduction in the mean CFUs/mL values and an increase in the mean bacterial load reduction percentages. The lowest mean post-irrigation CFUs/mL values were observed in the NaOCl + group, followed by NaOCl, PRO + , PRO, and saline groups respectively. Statistically significant differences were observed among all groups, except for the NaOCl and PRO + groups which did not exhibit any statistically significant difference. CONCLUSION Ultrasonically activated probiotic irrigant revealed an antibacterial effect similar to the conventional NaOCl and can be effectively used to fight against E. faecalis biofilm.
Collapse
Affiliation(s)
- Mai Sayed Hanafy
- Endodontic Department, Faculty of Dentistry, Suez University, Suez, Egypt.
| | - Noha Mohamed Kamal
- Botany Department, Faculty of Women for Arts, Science, and Education, Ain Shams University, Heliopolis, Egypt
| | - Hebatallah Atef Fathallah
- Endodontic Department, Faculty of Oral and Dental Medicine, Nahda University in Beni Suef, Beni Suef, Egypt
| |
Collapse
|
4
|
Gelen-Gungor D, Nigiz Ş, Özkul C, Eroğlu H, Nemutlu E, Ulubayram K, Mao Y, Michniak-Kohn B, Eroğlu İ. Co-delivery of Azithromycin and nisin through liposomes for skin infection to reduce antimicrobial drug resistance. Int J Pharm 2025:125764. [PMID: 40414326 DOI: 10.1016/j.ijpharm.2025.125764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/28/2025] [Accepted: 05/23/2025] [Indexed: 05/27/2025]
Abstract
Antimicrobial therapeutics are commonly used to treat infections caused by microorganisms. The standalone use of antimicrobial drugs frequently results in resistance development and reduced efficacy over prolonged treatments. Owing to funding challenges for developing novel drugs, one of the main European Medicines Agency activities to combat AMR is focusing on alternative therapies. Combination strategies integrating antimicrobial peptides with conventional drugs have demonstrated lower risk of resistance emergence and enhanced clinical efficacy. Nisin is generally recognized as safe peptide and has potential for clinical use. The aim of this study is to develop azithromycin and nisin-loaded liposome formulations for the topical treatment of skin infections that are both locally effective and capable of minimizing AMR. Liposomes were prepared by thin film hydration method, and homogeneous, stable, biocompatible liposomes demonstrated high encapsulation efficiencies (85 %). The optimized formulations were evaluated via in vitro release and ex vivo permeation studies by the Franz cell system. Approximately 250 μg/cm2 of AZM was released from combined formulations. The permeation through human cadaver skin and fluorescence imaging (Vertical Histological Cross-Section and Z-Stack) studies were performed to understand the performance and potential dermal applications of formulations. Labeled liposomes successfully localized within the skin layers by penetrating the stratum corneum. Antimicrobial activity and biofilm eradication against Staphylococcus aureus results showed that combined liposomal formulations were more effective than AZM-loaded formulations. In conclusion, this study presents co-delivery of AZM and nisin through liposome formulations as an effective treatment option for skin infection to reduce AMR.
Collapse
Affiliation(s)
- Dilek Gelen-Gungor
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Turkey; Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey
| | - Şeyma Nigiz
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, 06100, Sıhhiye, Ankara, Turkey
| | - Ceren Özkul
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, 06100, Sıhhiye, Ankara, Turkey
| | - Hakan Eroğlu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey; IPQ Pharma Medicines and Medical Devices Ltd. Co., 06800, Beytepe, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Department of Vaccine Technology, Vaccine Institute, Hacettepe University, Ankara, Turkey
| | - Kezban Ulubayram
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey; Department of Bioengineering, Hacettepe University, Ankara, Turkey
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Bozena Michniak-Kohn
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Center for Dermal Research, Life Sciences Building, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - İpek Eroğlu
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey; Department of Bioengineering, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
5
|
Garcia-Maset R, Chu V, Yuen N, Blumgart D, Yoon J, Murray BO, Joseph AA, Rohn JL. Effect of host microenvironment and bacterial lifestyles on antimicrobial sensitivity and implications for susceptibility testing. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:42. [PMID: 40399473 PMCID: PMC12095824 DOI: 10.1038/s44259-025-00113-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Bacterial infections remain a major global health issue, with antimicrobial resistance (AMR) worsening the crisis. However, treatment failure can occur even when bacteria show antibiotic susceptibility in diagnostic tests. We explore factors such as phenotypic resilience, bacterial lifestyles such as biofilms, and differences between laboratory tests and real infection sites, highlighting the need for improved platforms to better predict treatment outcomes, and reviewing emerging technologies aimed at improving susceptibility testing.
Collapse
Affiliation(s)
- Ramon Garcia-Maset
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Victoria Chu
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Nicholas Yuen
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Dalia Blumgart
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Jenny Yoon
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Benjamin O Murray
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Amelia A Joseph
- Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Jennifer L Rohn
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
6
|
Ciszkowicz E, Miłoś A, Łyskowski A, Buczkowicz J, Nieczaj A, Lecka-Szlachta K, Hus KK, Sikora K, Neubauer D, Bauer M, Kamysz W, Bocian A. AMPEC4: Naja ashei Venom-Derived Peptide as a Stimulator of Fibroblast Migration with Antibacterial Activity. Molecules 2025; 30:2167. [PMID: 40430339 PMCID: PMC12114029 DOI: 10.3390/molecules30102167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
The treatment of proctological conditions, including hemorrhoids, anal fissures, and perianal abscesses, is often complicated by bacterial infections, particularly those involving multidrug-resistant Escherichia coli. This study presents the synthesis, characterization, and biological evaluation of the newly designed synthetic peptide AMPEC4, inspired by cytotoxin 5 from Naja ashei snake venom. AMPEC4 demonstrated potent antimicrobial properties with MIC values of 100 and 200 µg/mL, effectively inhibiting biofilm formation (up to 84%) and eradicating the pre-formed biofilm by up to 35%. The antibacterial activity of AMPEC4 was further supported by a membrane permeabilization assay, demonstrating its capacity to disrupt bacterial membrane integrity in a dose-dependent manner. Furthermore, AMPEC4 significantly promoted fibroblast migration, a critical step in tissue regeneration, while exhibiting notable biocompatibility, as evidenced by the absence of hemolytic, cytotoxic, and genotoxic effects. By addressing both infection control and tissue regeneration, AMPEC4 represents a promising therapeutic strategy for managing chronic wounds, particularly in the challenging environment of the anorectal region. Its ability to target Escherichia coli reference and clinical strains while accelerating the wound-healing process underscores its potential for future clinical applications.
Collapse
Affiliation(s)
- Ewa Ciszkowicz
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| | - Anna Miłoś
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| | - Andrzej Łyskowski
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| | - Justyna Buczkowicz
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| | - Anna Nieczaj
- Doctoral School of the Rzeszów University of Technology, al. Powstańców Warszawy 12, 35-959 Rzeszów, Poland;
| | - Katarzyna Lecka-Szlachta
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| | - Konrad K. Hus
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| | - Karol Sikora
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (K.S.); (D.N.); (W.K.)
| | - Damian Neubauer
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (K.S.); (D.N.); (W.K.)
| | - Marta Bauer
- Department of Analytical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland;
| | - Wojciech Kamysz
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (K.S.); (D.N.); (W.K.)
| | - Aleksandra Bocian
- Department of Biotechnology and Bioinformatics, Faculty of Chemistry, Rzeszów University of Technology, al. Powstańców Warszawy 6, 35-959 Rzeszów, Poland; (A.M.); (J.B.); (K.L.-S.); (K.K.H.); (A.B.)
| |
Collapse
|
7
|
Aboelsaad E, Moustafa S, Amine A, Deghady A, El-Attar L. Platelet-rich plasma as a potential antimicrobial agent against multidrug-resistant bacteria in diabetic foot infections. Sci Rep 2025; 15:15145. [PMID: 40307308 PMCID: PMC12043966 DOI: 10.1038/s41598-025-97418-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Diabetes mellitus is a global public health concern, with diabetic foot infections (DFIs) being common clinical complications among affected patients. Bacterial isolates resistant to commonly used antimicrobial drugs are becoming more prevalent in DFIs. Some research suggests that platelet-rich plasma (PRP) may inhibit bacterial growth, making it a promising biological therapy. Therefore, an in vitro experimental study was conducted on 53 multidrug-resistant (MDR) bacterial strains isolated from DFIs. The isolates were methicillin-resistant Staphylococcus aureus (MRSA), MDR Klebsiella pneumoniae, and MDR Pseudomonas aeruginosa. The antibacterial activity of PRP was assessed using Kirby-Bauer disk diffusion method, broth microdilution method, checkerboard synergy testing, and time-kill assay. The time-kill assay demonstrated that PRP's antibacterial efficacy peaked during the second hour of incubation for MRSA and Pseudomonas aeruginosa, but peaked at the first hour for Klebsiella pneumoniae. However, the PPR's efficiency against all isolates decreased after the peak point, with no antibacterial activity observed at the 24th h of incubation. Additionally, biofilm inhibition and eradication assays revealed that PRP has no effect on biofilm formation. As a result, PRP has the ability to inhibit bacterial growth, although this effect is transient and depends on the bacterial strain.
Collapse
Affiliation(s)
- Engy Aboelsaad
- Department of Microbiology, High Institute of Public Health, Alexandria University, El-Horreya Road 165, Alexandria, 21561, Egypt.
| | - Sameh Moustafa
- Department of Vascular Surgery, Faculty of Medicine, Alexandria University, Chamblion Street, Alexandria, 21521, Egypt
| | - Amira Amine
- Department of Microbiology, High Institute of Public Health, Alexandria University, El-Horreya Road 165, Alexandria, 21561, Egypt
| | - Akram Deghady
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Chamblion Street, Alexandria, 21521, Egypt
| | - Laila El-Attar
- Department of Microbiology, High Institute of Public Health, Alexandria University, El-Horreya Road 165, Alexandria, 21561, Egypt
| |
Collapse
|
8
|
Galgano M, Pellegrini F, Mrenoshki D, Addante L, Sposato A, Del Sambro L, Capozzi L, Catalano E, Solito M, D’Amico F, Messina D, Parisi A, Pratelli A, Capozza P. Inhibition of Biofilm Production and Determination of In Vitro Time-Kill Thymus vulgaris L. Essential Oil (TEO) for the Control of Mastitis in Small Ruminants. Pathogens 2025; 14:412. [PMID: 40430732 PMCID: PMC12114205 DOI: 10.3390/pathogens14050412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Staphylococcus aureus and coagulase-negative staphylococci (CNS) are the main causative agents of mastitis in sheep. Their ability to form biofilms in vivo is considered an important virulence factor underlying mastitis outbreaks refractory to antibiotic treatments. Furthermore, pre- and postdipping immersion during milking in iodine substances could determine the presence of residues in milk and therefore represent a health risk factor for consumers. The aim of this study was to evaluate the antibacterial and biofilm inhibitory activity of Thymus vulgaris L. essential oil (TEO) against staphylococci strains isolated from ovine clinical mastitis. In particular, 3 reference strains (S. aureus 25923 and 11623 and S. epidermidis 12228) and 12 clinical isolates (6 S. aureus and 6 CNS) were used. TEO solutions, from a concentration of 1% (v/v) to 1.25% (v/v), corresponding to 9.28-2.32 mg/mL, were obtained after solubilization in 10% dimethyl sulfoxide (DMSO) and used to evaluate the bacterial time-kill compared to that of an iodine-based solution. Antibacterial efficacy was then assessed by the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC), while biofilm inhibition was assessed by minimum biofilm inhibitory concentration (MBIC) using a spectrophotometer at a wavelength of 570 nm. Additionally, biofilm-associated genes (icaA and icaD) were evaluated in all tested strains by PCR. The tested TEO concentrations were able to significantly and prominently reduce bacterial growth compared to controls, as demonstrated by bacterial time-kills. The MIC value was obtained at a concentration of 0.50% (v/v) for a single coagulation-positive isolate (S. aureus (f)) and at a concentration of 0.25% (v/v) for all other isolates. TEO showed effective bactericidal action with a 99.9% reduction in CFU/mL of all isolates in the MBC test at a concentration of 0.25% (v/v) for most of the tested strains. Furthermore, a marked inhibition in biofilm formation at all tested concentrations was observed, with MBIC value of 0.25%. All S. aureus tested were biofilm-producing strains and positive for icaA and icaD genes, while two CNS biofilm-producing strains were negative for both genes. These preliminary results suggest that TEO could be a promising alternative as an udder disinfectant during milking practices. Although in vivo studies are needed to confirm the efficacy and safety of TEO as an adjuvant in the prevention and treatment of udder infections, TEO could help counteract the emergence of antimicrobial resistance and reduce the potential risk of iodine residues in milk.
Collapse
Affiliation(s)
- Michela Galgano
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Francesco Pellegrini
- Department of Veterinary Medicine, University Aldo Moro of Bari, Sp Casamassima Km 3, Valenzano, 70010 Bari, Italy; (D.M.); (A.P.); (P.C.)
| | - Daniela Mrenoshki
- Department of Veterinary Medicine, University Aldo Moro of Bari, Sp Casamassima Km 3, Valenzano, 70010 Bari, Italy; (D.M.); (A.P.); (P.C.)
| | - Luciana Addante
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Alessio Sposato
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Carlo Forlanini, 27100 Pavia, Italy
| | - Laura Del Sambro
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Loredana Capozzi
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Elisabetta Catalano
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Marianna Solito
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Francesco D’Amico
- Istituto Zooprofilattico Sperimentale del Piemonte, Della Liguria e Della Valle d’Aosta, S.S. Genova e Portualità Marittima, 16129 Genova, Italy;
| | - Davide Messina
- Division of Veterinary Clinical Science, School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK;
| | - Antonio Parisi
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; (M.G.); (L.A.); (A.S.); (L.D.S.); (L.C.); (E.C.); (M.S.); (A.P.)
| | - Annamaria Pratelli
- Department of Veterinary Medicine, University Aldo Moro of Bari, Sp Casamassima Km 3, Valenzano, 70010 Bari, Italy; (D.M.); (A.P.); (P.C.)
| | - Paolo Capozza
- Department of Veterinary Medicine, University Aldo Moro of Bari, Sp Casamassima Km 3, Valenzano, 70010 Bari, Italy; (D.M.); (A.P.); (P.C.)
| |
Collapse
|
9
|
Yao L, Ma Y, Liu R, Li Y, Sun X, Wahafu T, Cao L, Mu W. Potential interactions between vancomycin and meropenem in culture-negative periprosthetic joint infection: an in vitro study. BMC Microbiol 2025; 25:197. [PMID: 40186104 PMCID: PMC11969832 DOI: 10.1186/s12866-025-03918-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Culture-negative periprosthetic joint infections (CN-PJIs) represent a critical subtype of PJI, and their high prevalence poses substantial challenges for treatment. CN-PJI is commonly managed utilizing antibiotic combinations, however, the interactions between these antibiotics have not been investigated. The aim of study was to investigate the synergistic and antagonistic effects of vancomycin (VAN) and meropenem (MEM), in an in vitro model of CN-PJI. METHODS Minimum Inhibitory Concentration (MIC), Minimum Bactericidal Concentration (MBC), Minimum Biofilm Inhibitory Concentration (MBIC), and Minimum Biofilm Eradication Concentration (MBEC) were determined for VAN and MEM. Fractional Inhibitory Concentration (FIC) and Fractional Biofilm Eradication Concentration (FBEC) indices were calculated to assess the synergistic or antagonistic effects of VAN in combination with MEM on Staphylococcus aureus (S. aureus), Staphylococcus epidermidis (S. epidermidis), and Escherichia coli (E. coli) incubated alone or in combination. RESULTS In the planktonic bacterial phase, MEM showed higher activity than VAN. Resistance increased when the bacteria were cultured together. The combination of VAN and MEM exhibited an indifferent effect against individual Staphylococci but an antagonistic effect against polymicrobial cultures. In biofilm, MEM demonstrated better antibiofilm activity than VAN, especially against E. coli biofilms. The combination of VAN and MEM showed an indifferent effect against E. coli and S. epidermidis-E. coli biofilms, but an antagonistic effect against S. aureus, S. epidermidis, S. aureus-S. epidermidis, and S. aureus-E. coli biofilms. CONCLUSION This study provides valuable insights into the effectiveness of VAN and MEM combinations in treating CN-PJIs, highlighting the need for careful consideration when selecting antibiotic treatments for these infections. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Liqin Yao
- Orthopedic Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Youcai Ma
- Department of Sports Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Rui Liu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 830054, Urumqi, China
| | - Yicheng Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 830054, Urumqi, China
| | - Xuebin Sun
- Department of Sports Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Tuerhongjiang Wahafu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 830054, Urumqi, China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 830054, Urumqi, China
| | - Wenbo Mu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 830054, Urumqi, China.
| |
Collapse
|
10
|
Wang D, Li Y, Li L, Chen Y, Min S, Wang Y, Feng J, Zhou J, Zhang Z, Fang Y. Synthesis of antibiofilm (1R,4S)-(-)-fenchone derivatives to control Pseudomonas syringae pv. tomato. PEST MANAGEMENT SCIENCE 2025; 81:1261-1273. [PMID: 39501917 DOI: 10.1002/ps.8525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/07/2024] [Accepted: 10/22/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND Biofilm plays a crucial role in Pseudomonas syringae pv. tomato (Pst) infection. We identified (1R,4S)-(-)-fenchone (FCH) as the most potent antibiofilm agent against Pst among 39 essential oil compounds. Subsequently, we synthesized a series of FCH oxime ester and acylhydrazine derivatives to explore more potent derivatives. RESULTS II3 was screened out as the most potent derivative, exhibiting a minimal biofilm inhibitory concentration of 60 μg mL-1 and a lowest concentration with maximal biofilm inhibition (LCMBI) of 200 μg mL-1, lower than those of FCH (80 and 500 μg mL-1, respectively). II3 and FCH showed minimum inhibitory concentration values >1000 μg mL-1 and similar maximal biofilm inhibition extents of 48.7% and 49.5% at their respective LCMBIs, respectively. Meanwhile, neither of them influenced cell viability or the activity of metabolic enzymes at their respective LCMBIs. II3 at its LCMBI significantly reduced biofilm thickness, extracellular polysaccharide content, and pectinase and cellulase production indices. In vivo assay results indicated that II3 could preventatively reduce the bacterial contents in tomato leaves at its LCMBI, and when combined with kasugamycin (KSG) (10 μg mL-1), II3 achieved the same level of bacterial reduction as the sole application of KSG (70 μg mL-1), thereby reducing the required dosage of KSG. Mechanistic studies demonstrated that II3 can down-regulate biofilm-related genes and inhibit PsyR/PsyI quorum sensing system, which differs from the bactericidal mechanisms. CONCLUSION These results underscore the potential of II3 as an antibiofilm agent for the control of Pst or FCH as a promising natural candidate for future in-depth optimization. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Delong Wang
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Yunpeng Li
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Linjing Li
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Yizhe Chen
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Shuoling Min
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Yong Wang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Juntao Feng
- State Key Laboratory of Crop Stress Biology for Arid Areas, Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Jianbo Zhou
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Zhijia Zhang
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Yali Fang
- College of Plant Protection, Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, China
| |
Collapse
|
11
|
Zafeer N, Mushtaq S, Shabbir S, Noor T, Imran M. Rhamnolipids functionalized intrinsically active liposomes loaded with cinnamaldehyde: Potent antimicrobial and antibiofilm activity against Salmonella Typhimurium and Salmonella Enteritidis. Food Sci Biotechnol 2025; 34:1063-1078. [PMID: 39974870 PMCID: PMC11832839 DOI: 10.1007/s10068-024-01735-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 02/21/2025] Open
Abstract
Intrinsically active rhamnolipids functionalized liposomes (rhamnosomes) as a carrier for cinnamaldehyde were developed to enhance the antibacterial activity against foodborne Salmonella isolates. Stable rhamnosomes were optimized with an excellent encapsulation efficiency of 85 ± 1%. SEM revealed slightly rough surface morphology and the mean diameter was slightly increased after the incorporation of cinnamaldehyde in rhamnosomes from 77 to 137 ± 2 nm. However, negative zeta-potential values were reduced from -18.2 ± 4 mV to - 13.4 ± 2 mV for cinnamaldehyde-loaded rhamnosomes. FTIR analyses revealed chemical interactions between rhamnolipids and phospholipids, which facilitated the development of rhamnosomes. Cinnamaldehyde-loaded rhamnosomes exhibited higher anti-Salmonella activity as compared to free cinnamaldehyde and void-rhamnosomes, and 75-80% reduction in biomass was observed due to their enhanced binding with the bacterial membrane in the antibiofilm assays. Antimicrobial results revealed that cinnamaldehyde-loaded rhamnosomes inhibited bacterial growth, displayed adequate biocompatibility and stability while providing an innovative strategy to control foodborne-resistant pathogens.
Collapse
Affiliation(s)
- Noureen Zafeer
- Department of Biosciences, COMSATS University Islamabad, Park Road, Islamabad, 45550 Pakistan
| | - Sajida Mushtaq
- SB Lab, SB Group of Companies, Murree Road, Rawalpindi, 46000 Pakistan
| | - Saima Shabbir
- Department of Materials Science and Engineering, Institute of Space Technology, Islamabad, 44000 Pakistan
| | - Tayyaba Noor
- School of Chemical and Materials Engineering (SCME), National University of Science and Technology (NUST), Islamabad, 44000 Pakistan
| | - Muhammad Imran
- Department of Biosciences, COMSATS University Islamabad, Park Road, Islamabad, 45550 Pakistan
| |
Collapse
|
12
|
Benny AT, Radhakrishnan EK. Assessing the antibiofilm activity of flavonol esters against Pseudomonas aeruginosa PAO1 biofilm: an in vitro, molecular docking, and molecular dynamics study. J Biomol Struct Dyn 2025; 43:813-829. [PMID: 39737751 DOI: 10.1080/07391102.2023.2283811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 10/24/2023] [Indexed: 01/01/2025]
Abstract
Pseudomonas aeruginosa is one of the opportunistic pathogens that may cause serious health problems and can produce several virulence factors, which are responsible for various infections, particularly in immunocompromised patients. They are responsible for producing infections on indwelling medical devices by attaching on to them and forming a biofilm. Antibiofilm, antivirulence, and gene expression studies of P. aeruginosa biofilm treated with esters of flavonols were evaluated. Pyocyanin, cell surface hydrophobicity, LasA protease estimation, rhamnolipid estimation, and pyoverdine estimation were performed to evaluate the antivirulence activities of the test compounds against P. aeruginosa. Previous studies on the antivirulence activity of flavonoids against P. aeruginosa demonstrate that even if they can inhibit bacterial growth, relatively high concentrations of the compound are generally required for the inhibition of virulence factors. The esters showed more than 40% inhibition in all the tested virulence factors at their sub minimum inhibitory concentration. The gene expression studies of selected esters toward lasB and rhlA genes show downregulation of rhlA which suggests the inhibition in biofilm formation through rhamnolipid inhibition, quorum sensing inhibition, or biofilm formation inhibition.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Anjitha Theres Benny
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, India
| | - Ethiraj K Radhakrishnan
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
13
|
Asadpour L, Mehrbakhsh Bandari MA, Sojoudi Masouleh R. Amikacin-loaded selenium nanoparticles improved antibacterial and antibiofilm activity of amikacin against bovine mastitis-causing Staphylococcus aureus. Heliyon 2025; 11:e41103. [PMID: 39758395 PMCID: PMC11699404 DOI: 10.1016/j.heliyon.2024.e41103] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background Antibiotic resistance in various microorganisms has become one of the most serious health problems worldwide. The use of nanoparticles in combination with conventional antibiotics is one of the recent efforts to overcome these challenges. This study aims to synthesize and evaluate the possibility of using amikacin-loaded selenium nanoparticles as antibacterial agent against multidrug-resistant Staphylococcus aureus, that causes bovine mastitis. Methods Selenium nanoparticles (SeNPs) were synthesized through chemical reduction of sodium selenite using L-cysteine. Loading of amikacin on selenium nanoparticles was done by mixing both in solution and confirmed by UV-Vis spectroscopy, XRD, SEM, and DLS. Antibacterial properties of obtained nanoparticles against S. aureus were determined using agar disc diffusion, broth micro dilution methods and also time-kill assay. Anti-biofilm properties of amikacin-loaded selenium nanoparticles was determined using microplate method and through determining the expression level of biofilm associated genes including icaA and icaD in S. aureus isolates treated with sub-MIC concentration of these nanoparticles by real-time PCR. Results Synthetized SeNPs and amikacin-loaded selenium nanoparticles (SeNPs@AMK) exhibited spherical appearances and 80.4 % of Se° had a diameter of 120 nm. SeNPs and SeNPs@AMK exhibited antibacterial effects against S. aureus isolates at the range of 32-128 μg/mL and 1-32 μg/mL respectively. Dependent on concentration and the exposure time, bacterial killing was promoted by the SeNPs@AMK treatment. The use of SeNPs@AMK decreased the biofilm formation of the isolates by more than 50 % and also lead to down-regulation of icaA and icaD biofilm associated gene compared to the control. Conclusions The results of this study suggest the antimicrobial properties of SeNPs and the reduction in the effective concentration of nanoparticle-loaded amikacin. Therefore, loading of antibiotics on the surface of nanoparticles may be used as a strategy to deal with the growing problem of drug resistance.
Collapse
Affiliation(s)
- Leila Asadpour
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | | | | |
Collapse
|
14
|
Bose S, Das SK. Biofilm Microenvironment-Sensitive Anti-Virulent and Immunomodulatory Nano-on-Nanodroplets to Combat Refractory Biofilm Infection Through Toxin Neutralization and Phagocytosis. Adv Healthc Mater 2025; 14:e2403528. [PMID: 39449220 DOI: 10.1002/adhm.202403528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/04/2024] [Indexed: 10/26/2024]
Abstract
Biofilm-associated wound infection is principally perceived as the bacterial defense mechanism that hinders antibiotic penetration, causes toxin impairment, and suppresses the immunological responses of the host immune system. Several antibiofilm agents have been developed, but the least of these agents can simultaneously cornerstone on the biofilm-associated immunosuppression and bacterial toxin-induced cellular dysfunction. Inspired by the fusogenic property of nanodroplets and immunomodulatory functions of metal nanoparticles, biofilm targeted anti-virulent immunomodulatory cationic nanoparticle shelled nanodroplets (C-AgND) is fabricated to completely disintegrate and eradicate the Staphylococcus aureus (S. aureus) biofilm. The specific binding of C-AgND neutralizes the negatively charged EPS layer, causing their destabilization followed by penetration of the nanoformulation into the biofilm matrix, killing the persister cells. Consequently, C-AgND eliminates the virulence property of the S. aureus biofilm through α-hemolysin neutralization. C-AgND promotes a strong immunomodulatory effect by polarizing macrophages into their M1 phenotype to induce phagocytosis of the disintegrated biofilm-released residual cells, rejuvenating the host's innate immune responses for the complete eradication of the biofilm. Moreover, the ex vivo skin wound infection model illustrates an excellent biofilm eradication efficacy of C-AgND in comparison to the commercial ones, rendering them to be a promising replacement of existing antibiofilm agents in clinical application.
Collapse
Affiliation(s)
- Somashree Bose
- Infectious Diseases and Immunology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sujoy K Das
- Infectious Diseases and Immunology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
15
|
Yin L, Guo Y, Xv X, Dai Y, Li L, Sun F, Lv X, Shu G, Liang X, He C, Xu Z, Ouyang P. Cinnamaldehyde nanoemulsion decorated with rhamnolipid for inhibition of methicillin-resistant Staphylococcus aureus biofilm formation: in vitro and in vivo assessment. Front Microbiol 2024; 15:1514659. [PMID: 39777149 PMCID: PMC11703839 DOI: 10.3389/fmicb.2024.1514659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) biofilm associated infections are prevalent and persistent, posing a serious threat to human health and causing significant economic losses in animal husbandry. Nanoemulsions demonstrate significant potential in the treatment of bacterial biofilm associated infections due to their unique physical, chemical and biological properties. In this study, a novel cinnamaldehyde nanoemulsion with the ability to penetrate biofilm structures and eliminate biofilms was developed. Methods The formulation of cinnamaldehyde nanoemulsion (Cin-NE) combined with rhamnolipid (RHL) was developed by self-assembly, and the efficacies of this formulation in inhibiting S. aureus biofilm associated infections were assessed through in vitro assays and in vivo experiments by a mouse skin wound healing model. Results The particle size of the selected Cin-NE formulation was 13.66 ± 0.08 nm, and the Cin-RHL-NE formulation was 20.45 ± 0.25 nm. The selected Cin-RHL-NE formulation was stable at 4, 25, and 37°C. Furthermore, the Minimum Inhibitory Concentration (MIC) value of Cin-RHL-NE against MRSA was two-fold lower than drug solution. Confocal laser scanning microscopy (CLSM) revealed the superior efficacy of Cin-RHL-NE in eradicating MRSA biofilms while maintaining the Cin's inherent functional properties. The efficacy of Cin-RHL-NE in the mouse skin wound healing model was superior to other formulation. Conclusion These findings highlight the potential of the formulation Cin-RHL-NE for eradicating biofilms, and effective in treating notoriously persistent bacterial infections. The Cin-RHL-NE can used as a dosage form of Cin application to bacterial biofilm associated infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhiwen Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, China
| |
Collapse
|
16
|
Nikolic I, Aleksic Sabo V, Gavric D, Knezevic P. Anti- Staphylococcus aureus Activity of Volatile Phytochemicals and Their Combinations with Conventional Antibiotics Against Methicillin-Susceptible S. aureus (MSSA) and Methicillin-Resistant S. aureus (MRSA) Strains. Antibiotics (Basel) 2024; 13:1030. [PMID: 39596725 PMCID: PMC11591321 DOI: 10.3390/antibiotics13111030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND MSSA and MRSA strains are challenging human pathogens that can develop resistance to antibiotics, highlighting the need for alternative antimicrobial agents. Plant metabolites, particularly volatile phytochemicals, may offer promising antimicrobial properties. The aim was to evaluate the antimicrobial and antibiofilm efficacy of various commercial volatile phytochemicals from the terpene and terpenoid groups against reference MSSA and MRSA strains, focusing on synergistic effects in both binary combinations and combinations with antibiotics. METHODS The microdilution method was used to determine the minimum inhibitory concentrations (MICs) for antibiotics and phytochemicals. The checkerboard method assessed synergistic interactions between phytochemicals and between phytochemicals and antibiotics, while the time-kill method was used to confirm these results. Biofilm quantification was performed using the microtiter plate method to evaluate the effects of phytochemicals, antibiotics, and their binary combinations on the eradication of 48-h-old biofilms. RESULTS Carvacrol and thymol demonstrated the strongest anti-staphylococcal activity, while other terpene compounds showed weaker effects. In binary combinations, carvacrol and thymol exhibited synergy against one MSSA strain (FICI = 0.50) and with tetracycline and chloramphenicol (FICI = 0.28-0.50). Synergy was also noted with streptomycin sulfate against one MRSA strain (FICI = 0.31-0.50) and with other antibiotics, including gentamicin (FICI = 0.25-0.50) and oxacillin (FICI = 0.44). Additionally, effective combinations achieved over 50% biofilm removal at both minimum inhibitory and sub-inhibitory concentrations. CONCLUSIONS Results showed that synergy varies based on strain sensitivity to chemical agents, highlighting their potential for personalized therapy. Despite the difficulty in removing preformed biofilms, the findings highlight the importance of combined treatments to enhance antibiotic effectiveness.
Collapse
Affiliation(s)
| | | | | | - Petar Knezevic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21000 Novi Sad, Serbia; (I.N.); (V.A.S.); (D.G.)
| |
Collapse
|
17
|
Madrid Sani AT, Ramos-Rocha KLV, Sarcinelli MA, Chaves MHDC, Rocha HVA, Léo P, Cerize NNP, Zanin MHA, Feitosa VA, Rangel-Yagui CDO. Development of a dry powder formulation for pulmonary delivery of azithromycin-loaded nanoparticles. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13635. [PMID: 39469425 PMCID: PMC11513329 DOI: 10.3389/jpps.2024.13635] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024]
Abstract
The COVID-19 pandemic has raised concern regarding respiratory system diseases and oral inhalation stands out as an attractive non-invasive route of administration for pulmonary diseases such as chronic bronchitis, cystic fibrosis, COVID-19 and community-acquired pneumonia. In this context, we encapsulated azithromycin in polycaprolactone nanoparticles functionalized with phospholipids rich in dipalmitoylphosphatidylcholine and further produced a fine powder formulation by spray drying with monohydrated lactose. Nanoparticles obtained by the emulsion/solvent diffusion-evaporation technique exhibited a mean hydrodynamic diameter around 195-228 nm with a narrow monomodal size distribution (PdI < 0.2). Nanoparticle dispersions were spray-dried at different inlet temperatures, atomizing air-flow, aspirator air flow, and feed rate, using lactose as a drying aid, resulting in a maximal process yield of 63% and an encapsulation efficiency of 83%. Excipients and the dry powder formulations were characterized in terms of morphology, chemical structure, thermal analyses and particle size by SEM, FTIR, DSC/TGA and laser light diffraction. The results indicated spherical particles with 90% at 4.06 µm or below, an adequate size for pulmonary delivery. Aerosolization performance in a NGI confirmed good aerodynamic properties. Microbiological assays showed that the formulation preserves AZM antimicrobial effect against Staphylococcus aureus and Streptococcus pneumoniae strains, with halos above 18 mm. In addition, no formulation-related cytotoxicity was observed against the human cell lines BEAS-2B (lung epithelial), HUVEC (endothelial) and HFF1 (fibroblasts). Overall, the approach described here allows the production of AZM-PCL nanoparticles incorporated into inhalable microparticles, enabling more efficient pulmonary therapy of lung infections.
Collapse
Affiliation(s)
- Alison Tatiana Madrid Sani
- Bionanomanufacturing Center, Technological Research Institute, São Paulo, Brazil
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo, São Paulo, Brazil
| | - Khellida Loiane V. Ramos-Rocha
- Bionanomanufacturing Center, Technological Research Institute, São Paulo, Brazil
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Patrícia Léo
- Bionanomanufacturing Center, Technological Research Institute, São Paulo, Brazil
| | | | | | - Valker Araujo Feitosa
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
18
|
Kim YH, Lee DH, Seo HS, Eun SH, Lee DS, Choi YK, Lee SH, Kim TY. Genome-based taxonomic identification and safety assessment of an Enterococcus strain isolated from a homemade dairy product. Int Microbiol 2024; 27:1513-1525. [PMID: 38466360 DOI: 10.1007/s10123-024-00496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
The aim of this study was to explore the taxonomic identification and evaluate the safety of a bacterium, Enterococcus lactis IDCC 2105, isolated from homemade cheese in Korea, using whole genome sequence (WGS) analysis. It sought to identify the species level of this Enterococcus spp., assess its antibiotic resistance, and evaluate its virulence potential. WGS analysis confirmed the bacterial strain IDCC 2105 as E. lactis and identified genes responsible for resistance to erythromycin and clindamycin, specifically msrC, and eatAv, which are chromosomally located, indicating a minimal risk for horizontal gene transfer. The absence of plasmids in E. lactis IDCC 2105 further diminishes the likelihood of resistance gene dissemination. Additionally, our investigation into seven virulence factors, including hemolysis, platelet aggregation, biofilm formation, hyaluronidase, gelatinase, ammonia production, and β-glucuronidase activity, revealed no detectable virulence traits. Although bioinformatic analysis suggested the presence of collagen adhesion genes acm and scm, these were not corroborated by phenotypic virulence assays. Based on these findings, E. lactis IDCC 2105 presents as a safe strain for potential applications, contributing valuable information on its taxonomy, antibiotic resistance profile, and lack of virulence factors, supporting its use in food products.
Collapse
Affiliation(s)
- Young-Hoo Kim
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, South Korea
| | | | - Han Sol Seo
- Yunovia Co., Ltd, Hwaseong, 18449, South Korea
| | | | - Do Sup Lee
- Yunovia Co., Ltd, Hwaseong, 18449, South Korea
| | | | - Sang Hyun Lee
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, South Korea
| | - Tae-Yoon Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, 13488, Republic of Korea.
| |
Collapse
|
19
|
Carmona-Orozco ML, Echeverri F. Induction of biofilm in extended-spectrum beta-lactamase Staphylococcus aureus with drugs commonly used in pharmacotherapy. Microb Pathog 2024; 195:106863. [PMID: 39159772 DOI: 10.1016/j.micpath.2024.106863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Staphylococcus aureus is a bacterial pathogen that causes bloodstream infections, pneumonia, and skin abscesses and is the primary pathogen responsible for medical devices associated with biofilm infections, accounting for approximately 70 % of cases. Therefore, the World Health Organization (WHO) has designated this microorganism as a top priority due to its role in causing over 20,000 bacteremia-related deaths in the US each year. The issue of pathogen resistance to antibiotics, mainly by a biofilm, further complicates these infections since biofilms render the bacterial colony impervious to antibiotics. However, many natural and synthetic substances also induce bacterial biofilm formation. Therefore, we investigated whether the most common active pharmaceutical ingredients (APIs) could induce biofilm formation in two clinical isolates of extended-spectrum beta-lactamase Staphylococcus aureus, one of them also methicillin-resistant (A2M) and two medical devices. We detected biofilm inducers, inhibitors, and destabilizers. Microbial strain, medical devices, API structure, and concentration influenced the modulatory effects of biofilm. In all devices tested, including microplates, FR18 duodenal probe, and respiratory probe, the clinic isolate methicillin-resistant S. aureus A2M exhibited lower susceptibility to biofilm formation than S. aureus A1. The anti-inflammatory acetaminophen, the hypocholesterolemic lovastatin, and the diuretic hydrochlorothiazide all induced biofilm. However, verapamil, an antihypertensive, and cetirizine, an antihistamine, inhibited biofilm on S. aureus A2M, while propranolol, another antihypertensive, inhibited biofilm on S. aureus A1. Additionally, diclofenac, an analgesic, and cetirizine destabilized the biofilm, resulting in more free bacteria and possibly making them more susceptible to external agents such as antibiotics. Nonetheless, further epidemiologic analyses and in vivo assays are needed to confirm these findings and to establish a correlation between drug use, the onset of bacterial infections in patients, and the use of medical devices. This work provides information about the probable clinical implications of drugs in patients using medical devices or undergoing surgical procedures. Inhibitory APIs could also be used as drug repurposing or templates to design new, more potent biofilm inhibitors.
Collapse
Affiliation(s)
- Maria L Carmona-Orozco
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia
| | - Fernando Echeverri
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
20
|
Serrano S, Ferreira MV, Alves-Barroco C, Morais S, Barreto-Crespo MT, Tenreiro R, Semedo-Lemsaddek T. Beyond Harmful: Exploring Biofilm Formation by Enterococci Isolated from Portuguese Traditional Cheeses. Foods 2024; 13:3067. [PMID: 39410102 PMCID: PMC11476095 DOI: 10.3390/foods13193067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
This study investigated the biofilm-forming capabilities of Enterococcus isolates from Portuguese traditional cheeses with protected designation of origin (PDO) status, specifically Azeitão and Nisa. Given the absence of added starter cultures in the cheesemaking process, the characteristics of these cheeses are intrinsically linked to the autochthonous microbiota present in the raw materials and the production environment. Our findings demonstrate that all isolates possess biofilm production abilities, which are crucial for their colonization and persistence within cheese factories, thereby maintaining factory-specific microbial heritage. Through an integrated analysis utilizing principal component analysis (PCA), a direct correlation between biofilm formation and cell viability was established. Notably, these results underscore the adaptive capacity of enterococci to survive environmental fluctuations and their role in the unique characteristics of Portuguese traditional cheeses. Overall, this research enhances our understanding of the microbial dynamics in cheese production and highlights the importance of enterococci in preserving cheese quality and heritage.
Collapse
Affiliation(s)
- Susana Serrano
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (S.S.); (S.M.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| | | | - Cinthia Alves-Barroco
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (S.S.); (S.M.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| | - Susana Morais
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (S.S.); (S.M.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| | - Maria Teresa Barreto-Crespo
- iBET, Institute of Experimental Biology and Technology, P.O. Box 12, 2781-901 Oeiras, Portugal;
- ITQB, Institute of Chemical and Biological Technology António Xavier, Nova University of Lisbon, Republic Avenue, 2780-157 Oeiras, Portugal
| | - Rogério Tenreiro
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
| | - Teresa Semedo-Lemsaddek
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; (S.S.); (S.M.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- BioISI—Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
| |
Collapse
|
21
|
Mahmoud GAE, Rashed NM, El-Ganainy SM, Salem SH. Unveiling the Neem ( Azadirachta indica) Effects on Biofilm Formation of Food-Borne Bacteria and the Potential Mechanism Using a Molecular Docking Approach. PLANTS (BASEL, SWITZERLAND) 2024; 13:2669. [PMID: 39339644 PMCID: PMC11434743 DOI: 10.3390/plants13182669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
Biofilms currently represent the most prevalent bacterial lifestyle, enabling them to resist environmental stress and antibacterial drugs. Natural antibacterial agents could be a safe solution for controlling bacterial biofilms in food industries without affecting human health and environmental safety. A methanolic extract of Azadirachta indica (neem) leaves was prepared and analyzed using gas chromatography-mass spectrometry for the identification of its phytochemical constituents. Four food-borne bacterial pathogens (Bacillus cereus, Novosphingobium aromaticivorans, Klebsiella pneumoniae, and Serratia marcescens) were tested for biofilm formation qualitatively and quantitatively. The antibacterial and antibiofilm properties of the extract were estimated using liquid cultures and a microtiter plate assay. The biofilm inhibition mechanisms were investigated using a light microscope and molecular docking technique. The methanolic extract contained 45 identified compounds, including fatty acids, ester, phenols, flavonoids, terpenes, steroids, and antioxidants with antimicrobial, anticancer, and anti-inflammatory properties. Substantial antibacterial activity in relation to the extract was recorded, especially at 100 μg/mL against K. pneumoniae and S. marcescens. The extract inhibited biofilm formation at 100 μg/mL by 83.83% (S. marcescens), 73.12% (K. pneumoniae), and 54.4% (N. aromaticivorans). The results indicate efficient biofilm formation by the Gram-negative bacteria S. marcescens, K. pneumoniae, and N. aromaticivorans, giving 0.74, 0.292, and 0.219 OD at 595 nm, respectively, while B. cereus was found to have a low biofilm formation potential, i.e., 0.14 OD at 595 nm. The light microscope technique shows the antibiofilm activities with the biofilm almost disappearing at 75 μg/mL and 100 μg/mL concentrations. This antibiofilm property was attributed to DNA gyrase inhibition as illustrated by the molecular docking approach.
Collapse
Affiliation(s)
| | - Nahed M Rashed
- Department of Arid Land Agriculture, College of Agricultural and Food Sciences, King Faisal University, P.O. Box 420, Al-Ahsa 31982, Saudi Arabia
- Horticulture Department, Faculty of Agriculture, Damietta University, Damietta 34519, Egypt
| | - Sherif M El-Ganainy
- Department of Arid Land Agriculture, College of Agricultural and Food Sciences, King Faisal University, P.O. Box 420, Al-Ahsa 31982, Saudi Arabia
- Plant Pathology Research Institute, Agricultural Research Center, Giza 12619, Egypt
| | - Shimaa H Salem
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
22
|
Barta A, Salusso A, Kúsz N, Berkecz R, Schlauer J, Purger D, Hohmann J, Carpinella MC, Vasas A. Phenanthrenes from Juncus articulatus with Antibacterial and Biofilm Formation Inhibitory Activity. JOURNAL OF NATURAL PRODUCTS 2024; 87:2068-2080. [PMID: 39121346 PMCID: PMC11348428 DOI: 10.1021/acs.jnatprod.4c00577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 08/11/2024]
Abstract
Continuing our search for bioactive compounds in species from the Juncaceae family, Juncus articulatus was investigated. Ten previously undescribed phenanthrenes─articulins A-J (1-10)─and ten known compounds─juncuenin B, dehydrojuncuenin B, juncatrin B, ensifolins E, F, H, I, K, juncuenin D, and luzulin A (11-20)─along with other compounds, have been isolated and identified. The isolated compounds were evaluated for antibacterial activity against Escherichia coli, Pseudomonas aeruginosa, methicillin-susceptible Staphylococcus aureus (MSSA), and methicillin-resistant Staphylococcus aureus (MRSA). Compounds 12 and 14 exhibited the most potent activity against planktonic and sessile MSSA and MRSA with minimum inhibitory concentration (MIC) values of 15.1 μM (12 for both bacterial strains) and 15.3 μM (14 for both bacterial strains). Compounds 15, 17, and 18 also exhibited activity against both strains, although to a lower extent, with MIC values ranging from 30.0 to 56.8 μM. The inhibition of biofilm formation of these compounds was observed at 15.1-114.3 μM. This study elucidates the phenanthrene composition of J. articulatus and the antibacterial effect of these compounds.
Collapse
Affiliation(s)
- Anita Barta
- Institute
of Pharmacognosy, University of Szeged, Szeged 6720, Hungary
- HUN-REN-USZ
Biologically Active Natural Products Research Group, University of Szeged, Szeged 6720, Hungary
| | - Agostina Salusso
- Fine
Chemical and Natural Products Laboratory, CIDIE CONICET-UCC, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina
| | - Norbert Kúsz
- Institute
of Pharmacognosy, University of Szeged, Szeged 6720, Hungary
| | - Róbert Berkecz
- Institute
of Pharmaceutical Analysis, University of
Szeged, Szeged 6720, Hungary
| | - Jan Schlauer
- The
Center for Plant Molecular Biology, University
of Tubingen, Tubingen 72076, Germany
| | - Dragica Purger
- Department
of Pharmacognosy, University of Pécs, Pécs 7624, Hungary
| | - Judit Hohmann
- Institute
of Pharmacognosy, University of Szeged, Szeged 6720, Hungary
- HUN-REN-USZ
Biologically Active Natural Products Research Group, University of Szeged, Szeged 6720, Hungary
| | - Maria Cecilia Carpinella
- Fine
Chemical and Natural Products Laboratory, CIDIE CONICET-UCC, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina
| | - Andrea Vasas
- Institute
of Pharmacognosy, University of Szeged, Szeged 6720, Hungary
- HUN-REN-USZ
Biologically Active Natural Products Research Group, University of Szeged, Szeged 6720, Hungary
| |
Collapse
|
23
|
Saoudi B, Bariz K, Saci S, Belounis Y, Ait Issad H, Abbaci M, Mustapha MA, Nabti EH, Alenazy R, Alhussaini MS, Alyahya AAI, Alqasmi M, Alhumaidi MS, Almufarriji FM, Houali K. Enhancing Antibiotic Efficacy and Combating Biofilm Formation: Evaluating the Synergistic Potential of Origanum vulgare Essential Oil against Multidrug-Resistant Gram-Negative Bacteria. Microorganisms 2024; 12:1651. [PMID: 39203493 PMCID: PMC11356740 DOI: 10.3390/microorganisms12081651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Multidrug-resistant (MDR) Gram-negative bacteria remain a global public health issue due to the barrier imposed by their outer membrane and their propensity to form biofilms. It is becoming imperative to develop new antibacterial strategies. In this context, this study aims to evaluate the antibacterial efficacy of Origanum vulgare essential oil (OEO), alone and in combination with antibiotics, as well as its antibiofilm action against multidrug-resistant Gram-negative strains. OEO components were identified by gas chromatography-mass spectrometry (GC-MS), and antibacterial activity was assessed using the agar diffusion test and the microdilution method. Interactions between OEO and antibiotics were examined using the checkerboard method, while antibiofilm activity was analyzed using the crystal violet assay. Chemical analysis revealed that carvacrol was the major compound in OEO (61.51%). This essential oil demonstrated activity against all the tested strains, with inhibition zone diameters (IZDs) reaching 32.3 ± 1.5 mm. The combination of OEO with different antibiotics produced synergistic and additive effects, leading to a reduction of up to 98.44% in minimum inhibitory concentrations (MICs). In addition, this essential oil demonstrated an ability to inhibit and even eradicate biofilm formation. These results suggest that OEO could be exploited in the development of new molecules, combining its metabolites with antibiotics.
Collapse
Affiliation(s)
- Bilal Saoudi
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Karim Bariz
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Sarah Saci
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Yousra Belounis
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Hakima Ait Issad
- Laboratoire Ressources Naturelles, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria;
| | - Mohamed Abbaci
- Centre de Recherche Scientifique et Technique en Analyses Physico—Chimiques CRAPC, Bou Ismail 42004, Algeria; (M.A.); (M.A.M.)
| | - Mohamed Abou Mustapha
- Centre de Recherche Scientifique et Technique en Analyses Physico—Chimiques CRAPC, Bou Ismail 42004, Algeria; (M.A.); (M.A.M.)
| | - El-Hafid Nabti
- Laboratoire de Maitrise des Energies Renouvelables, Faculté des Sciences de la Nature et de le Vie, Université de Bejaïa, Bejaïa 06000, Algeria;
| | - Rawaf Alenazy
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Mohammed Sanad Alhussaini
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Abdulrahman A. I. Alyahya
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Mohammed Alqasmi
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Maryam S. Alhumaidi
- Department of Biology, College of Science, University of Hafr Al Batin, P.O. Box 1803, Hafr Al Batin 31991, Saudi Arabia;
| | - Fawaz M. Almufarriji
- Medical Laboratories Department, College of Applied Medical Sciences in Al-Quwayiyah, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Karim Houali
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| |
Collapse
|
24
|
Negi M, Kaushik N, Lamichhane P, Jaiswal A, Borkar SB, Patel P, Singh P, Choi EH, Kaushik NK. Biocompatible plasma-treated liquids: A sustainable approach for decontaminating gastrointestinal-infection causing pathogens. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134562. [PMID: 38743977 DOI: 10.1016/j.jhazmat.2024.134562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Nosocomial infections are a serious threat and difficult to cure due to rising antibiotic resistance in pathogens and biofilms. Direct exposure to cold atmospheric plasma (CAP) has been widely employed in numerous biological research endeavors. Nonetheless, plasma-treated liquids (PTLs) formulated with physiological solutions may offer additional benefits such as enhanced portability, and biocompatibility. Additionally, CAP-infused long-lived reactive oxygen and nitrogen species (RONS) such as nitrite (NO2-), nitrate (NO3-), and hydrogen peroxide (H2O2) can synergistically induce their antibacterial activity. Herein, we investigated those argon-plasma jet-treated liquids, including Ringer's lactate (RL), phosphate-buffered saline (PBS), and physiological saline, have significant antibacterial activity against nosocomial/gastrointestinal-causing pathogens, which might be due to ROS-mediated lipid peroxidation. Combining the conventional culture-based method with propidium iodide monoazide quantitative PCR (PMAxx™-qPCR) indicated that PTLs induce a minimal viable but non-culturable (VBNC) state and moderately affect culturable counts. Specifically, the PTL exposure resulted in pathogenicity dysfunction via controlling T3SS-related effector genes of S. enterica. Overall, this study provides insights into the effectiveness of PTLs for inducing ROS-mediated damage, controlling the virulence of diarrheagenic bacteria, and modulating homeostatic genes.
Collapse
Affiliation(s)
- Manorma Negi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, South Korea.
| | - Prajwal Lamichhane
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea
| | - Apurva Jaiswal
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea
| | - Shweta B Borkar
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea
| | - Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea
| | - Prashant Singh
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Delhi, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, South Korea.
| |
Collapse
|
25
|
Prosswimmer T, Nick SE, Bryers JD, Daggett V. Designed De Novo α-Sheet Peptides Destabilize Bacterial Biofilms and Increase the Susceptibility of E. coli and S. aureus to Antibiotics. Int J Mol Sci 2024; 25:7024. [PMID: 39000131 PMCID: PMC11241457 DOI: 10.3390/ijms25137024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Biofilm-associated microbes are 10-1000 times less susceptible to antibiotics. An emerging treatment strategy is to target the structural components of biofilm to weaken the extracellular matrix without introducing selective pressure. Biofilm-associated bacteria, including Escherichia coli and Staphylococcus aureus, generate amyloid fibrils to reinforce their extracellular matrix. Previously, de novo synthetic α-sheet peptides designed in silico were shown to inhibit amyloid formation in multiple bacterial species, leading to the destabilization of their biofilms. Here, we investigated the impact of inhibiting amyloid formation on antibiotic susceptibility. We hypothesized that combined administration of antibiotics and α-sheet peptides would destabilize biofilm formation and increase antibiotic susceptibility. Two α-sheet peptides, AP90 and AP401, with the same sequence but inverse chirality at every amino acid were tested: AP90 is L-amino acid dominant while AP401 is D-amino acid dominant. For E. coli, both peptides increased antibiotic susceptibility and decreased the biofilm colony forming units when administered with five different antibiotics, and AP401 caused a greater increase in all cases. For S. aureus, increased biofilm antibiotic susceptibility was also observed for both peptides, but AP90 outperformed AP401. A comparison of the peptide effects demonstrates how chirality influences biofilm targeting of gram-negative E. coli and gram-positive S. aureus. The observed increase in antibiotic susceptibility highlights the role amyloid fibrils play in the reduced susceptibility of bacterial biofilms to specific antibiotics. Thus, the co-administration of α-sheet peptides and existing antibiotics represents a promising strategy for the treatment of biofilm infections.
Collapse
Affiliation(s)
- Tatum Prosswimmer
- Molecular Engineering Program, University of Washington, Seattle, WA 98195, USA
| | - Sarah E. Nick
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| | - James D. Bryers
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| | - Valerie Daggett
- Molecular Engineering Program, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
26
|
Polat T, Soyhan İ, Cebeci S, İldeniz TAÖ, Gök Ö, Elmas MA, Mozioğlu E, Ünübol N. New-generation biofilm effective antimicrobial peptides and a real-time anti-biofilm activity assay: CoMIC. Appl Microbiol Biotechnol 2024; 108:316. [PMID: 38700735 PMCID: PMC11068671 DOI: 10.1007/s00253-024-13134-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 05/06/2024]
Abstract
Nowadays, it is very important to produce new-generation drugs with antimicrobial properties that will target biofilm-induced infections. The first target for combating these microorganisms, which are the source itself. Antimicrobial peptides, which are more effective than antibiotics due to their ability to kill microorganisms and use a different metabolic pathway, are among the new options today. The aim of this study is to develop new-generation antibiotics that inhibit both biofilm-producing bacteria and the biofilm itself. For this purpose, we designed four different peptides by combining two amino acid forms (D- and L-) with the same sequence having alpha helix structures. It was found that the combined use of these two forms can increase antimicrobial efficacy more than 30-fold. These results are supported by molecular modeling and scanning electron microscopy (SEM), at the same time cytotoxicity (IC50) and hemotoxicity (HC50) values remained within the safe range. Furthermore, antibiofilm activities of these peptides were investigated. Since the existing biofilm inhibition methods in the literature do not technically simulate the exact situation, in this study, we have developed a real-time observable biofilm model and a new detection method based on it, which we call the CoMIC method. Findings have shown that the NET1 peptide with D-leucine amino acid in its structure and the NET3 peptide with D-arginine amino acid in its structure are effective in inhibiting biofilm. As a conclusion, our peptides can be considered as potential next-generation broad-spectrum antibiotic molecule/drug candidates that might be used in biofilm and clinical important bacteria. KEY POINTS: • Antimicrobial peptides were developed to inhibit both biofilms producing bacteria and the biofilm itself. • CoMIC will fill a very crucial gap in understanding biofilms and conducting the necessary quantitative studies. • Molecular modelling studies, NET1 peptide molecules tends to move towards and adhere to the membrane within nanoseconds.
Collapse
Affiliation(s)
- Tuba Polat
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - İrem Soyhan
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Sinan Cebeci
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Tuğba Arzu Özal İldeniz
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Özgül Gök
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Merve Açıkel Elmas
- Department of Histology and Embriology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Erkan Mozioğlu
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| | - Nihan Ünübol
- Department of Medical Microbiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
- Medical Laboratory Techniques, Vocational School of Health Services, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| |
Collapse
|
27
|
Wang C, Defoirdt T, Rajkovic A. The impact of indole and mucin on sporulation, biofilm formation, and enterotoxin production in foodborne Clostridium perfringens. J Appl Microbiol 2024; 135:lxae083. [PMID: 38544331 DOI: 10.1093/jambio/lxae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/13/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024]
Abstract
AIMS Indole and mucin are compounds found in the host environment as they are produced by the host or by the host-associated microbiota. This study investigated whether indole and mucin impact Clostridium perfringens growth and sporulation, as well as enterotoxin production and biofilm formation. METHODS AND RESULTS There was no impact on growth of Cl. perfringens for up to 400 µM indole and 240 mg/l mucin, and neither indole nor mucin affected sporulation. Reverse-transcriptase qPCR showed that mucin strongly upregulated the expression of Cl. perfringens enterotoxin (up to 121-fold increase), whereas indole had a much more modest effect (2-fold). This was also reflected in increased Cl. perfringens enterotoxin levels in mucin-treated Cl. perfringens (as assessed by a reversed passive latex agglutination assay). Finally, mucin and indole significantly increased biofilm formation of Cl. perfringens, although the effect size was relatively small (less than 1.5 fold). CONCLUSION These results indicate that Cl. perfringens can sense its presence in a host environment by responding to mucin, and thereby markedly increased enterotoxin production.
Collapse
Affiliation(s)
- Chao Wang
- Research Unit Food Microbiology and Food Preservation, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Tom Defoirdt
- Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Andreja Rajkovic
- Research Unit Food Microbiology and Food Preservation, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| |
Collapse
|
28
|
Hussan, Nisa S, Bano SA, Zia M. Chemically synthesized ciprofloxacin-PEG-FeO nanotherapeutic exhibits strong antibacterial and controlled cytotoxic effects. Nanomedicine (Lond) 2024; 19:875-893. [PMID: 38530883 DOI: 10.2217/nnm-2023-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024] Open
Abstract
Aim: To develop a biocompatible conjugated ciprofloxacin-PEG-FeO nanodelivery system with increased efficacy of available therapeutics in a controlled manner. Materials & methods: FeO nanoparticles were synthesized by chemical and biological methods and modified as ciprofloxacin-PEG-FeO nanoformulations. After initial antibacterial and cytotoxicity studies, the effective and biocompatible nanoformulations was further fabricated as nanotherapeutics for in vivo studies in mouse models. Results: Chemically synthesized ciprofloxacin-PEG-FeO nanoformulations demonstrated boosted antibacterial activity against clinically isolated bacterial strains. Nanoformulations were also found to be compatible with baby hamster kidney 21 cells and red blood cells. In in vivo studies, nanotherapeutic showed wound-healing effects with eradication of Staphylococcus aureus infection. Conclusion: The investigations indicate that the developed nanotherapeutic can eradicate localized infections and enhance wound healing with controlled cytotoxicity.
Collapse
Affiliation(s)
- Hussan
- Department of Microbiology, University of Haripur, Khyber Pakhtunkhwa, 22620, Pakistan
| | - Sobia Nisa
- Department of Microbiology, University of Haripur, Khyber Pakhtunkhwa, 22620, Pakistan
| | - Syeda Asma Bano
- Department of Microbiology, University of Haripur, Khyber Pakhtunkhwa, 22620, Pakistan
| | - Muhammad Zia
- Department of Biotechnology, Quaid e Azam University Islamabad, Islamabad, 15320, Pakistan
| |
Collapse
|
29
|
Cherian LM, Liu S, Hon K, Ramezanpour M, Hu H, Javadiyan S, Stafford I, Wormald PJ, Psaltis AJ, Vreugde S. Hydrocortisone metabolism by Staphylococcus: Effects on anti-inflammatory and antimicrobial action. Int Forum Allergy Rhinol 2024; 14:711-715. [PMID: 37466463 DOI: 10.1002/alr.23242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/20/2023]
Abstract
KEY POINTS Hydrocortisone 21-hemisuccinate (HCHS) influenced the growth and metabolism of Staphylococcus aureus S. aureus metabolic activity was high and antibiotic susceptibility low at 1.4 mg/mL HCHS S. aureus metabolized HCHS to cortisol and reduced poly(I:C)-induced IL-6 secretion.
Collapse
Affiliation(s)
- Lisa Mary Cherian
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
- Department of Otolaryngology, Head and Neck Surgery, Christian Medical College, Vellore, Tamil Nadu, India
| | - Sha Liu
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Karen Hon
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Mahnaz Ramezanpour
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Hua Hu
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Shari Javadiyan
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Irene Stafford
- Department of Cardiology, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Peter-John Wormald
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Alkis James Psaltis
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Sarah Vreugde
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| |
Collapse
|
30
|
Klopper KB, Bester E, van Schalkwyk M, Wolfaardt GM. Highlighting the limitations of static microplate biofilm assays for industrial biocide effectiveness compared to dynamic flow conditions. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e13214. [PMID: 38015101 PMCID: PMC10866068 DOI: 10.1111/1758-2229.13214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023]
Abstract
The minimal inhibitory concentration of an antimicrobial required to inhibit the growth of planktonic populations (minimum inhibitory concentration [MIC]) remains the 'gold standard' even though biofilms are acknowledged to be recalcitrant to concentrations that greatly exceed the MIC. As a result, most studies focus on biofilm tolerance to high antimicrobial concentrations, whereas the effect of environmentally relevant sub-MIC on biofilms is neglected. The effect of the MIC and sub-MIC of an isothiazolinone biocide on a microbial community isolated from an industrial cooling system was assessed under static and flow conditions. The differential response of planktonic and sessile populations to these biocide concentrations was discerned by modifying the broth microdilution assay. However, the end-point analysis of biofilms cultivated in static microplates obscured the effect of sub-MIC and MIC on biofilms. A transition from batch to the continuous flow system revealed a more nuanced response of biofilms to these biocide concentrations, where biofilm-derived planktonic cell production was maintained despite an increase in the frequency and extent of biofilm sloughing. A holistic, 'best of both worlds' approach that combines the use of static and continuous flow systems is useful to investigate the potential for the development of persistent biofilms under conditions where exposure to sub-MIC and MIC may occur.
Collapse
Affiliation(s)
- Kyle B. Klopper
- Department of MicrobiologyStellenbosch UniversityStellenboschSouth Africa
| | - Elanna Bester
- Department of MicrobiologyStellenbosch UniversityStellenboschSouth Africa
| | | | - Gideon M. Wolfaardt
- Department of MicrobiologyStellenbosch UniversityStellenboschSouth Africa
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| |
Collapse
|
31
|
El-Newehy MH, Aldalbahi A, Thamer BM, Hameed MMA. Establishment and inactivation of mono-species biofilm in a semipilot-scale water distribution system using nanocomposite of silver nanoparticles/montmorillonite loaded cationic chitosan. Int J Biol Macromol 2024; 258:128874. [PMID: 38128797 DOI: 10.1016/j.ijbiomac.2023.128874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 12/10/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023]
Abstract
This study presents a novel approach in the synthesis and characterization of nanocomposites comprising cationic chitosan (CCS) blended with varying concentrations of silver nanoparticles/montmorillonite (AgNPs/MMT). AgNPs/MMT was synthesized using soluble starch as a reducing and stabilizing agent. Subsequently, nanocomposites, namely CCS/AgMMT-0, CCS/AgMMT-0.5, CCS/AgMMT-1.5, and CCS/AgMMT-2.5, were developed by blending 2.5 g of CCS with 0, 0.5, 1.5, and 2.5 g of AgNPs/MMT, respectively, and the corresponding nanocomposites were prepared using ball milling technique. Transmission electron microscopy (TEM) analysis revealed the formation of nanocomposites that exhibiting nearly spherical morphologies. Dynamic light scattering (DLS) measurements displayed average particle sizes of 1183 nm, 131 nm, 140 nm, and 188 nm for CCS/AgMMT-0, CCS/AgMMT-0.5, CCS/AgMMT-1.5, and CCS/AgMMT-2.5, respectively. The narrow polydispersity index (~0.5) indicated uniform particle size distributions across the nanocomposites, affirming monodispersity. Moreover, the zeta potential values exceeding 30 mV across all nanocomposites that confirmed their stability against agglomeration. Notably, CCS/AgMMT-2.5 nanocomposite exhibited potent antibacterial and antibiofilm properties against diverse pipeline materials. Findings showed that after 15 days of incubation, the highest populations of biofilm cells, Pseudomonas aeruginosa biofilm, developed over UPVC, MDPE, DCI, and SS, with corresponding HPCs of 4.79, 6.38, 8.81, and 7.24 CFU/cm2. The highest cell densities of Enterococcus faecalis biofilm in the identical situation were 4.19, 5.89, 8.12, and 6.9 CFU/cm2. The nanocomposite CCS/AgMMT-2.5 exhibited the largest measured zone of inhibition (ZOI) against both P. aeruginosa and E. faecalis, with measured ZOI values of 19 ± 0.65 and 17 ± 0.21 mm, respectively. Remarkably, the research indicates that the youngest biofilm exhibited the most notable rate of inactivation when exposed to a dose of 150 mg/L, in comparison to the mature biofilm. These such informative findings could offer valuable insights into the development of effective antibiofilm agents and materials applicable in diverse sectors such as water treatment facilities, medical devices, and industrial pipelines.
Collapse
Affiliation(s)
- Mohamed H El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Ali Aldalbahi
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Badr M Thamer
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Meera Moydeen Abdul Hameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
32
|
Guliy OI, Evstigneeva SS, Shirokov AA, Bunin VD. Sensor system for analysis of biofilm sensitivity to ampicillin. Appl Microbiol Biotechnol 2024; 108:172. [PMID: 38265501 PMCID: PMC10808281 DOI: 10.1007/s00253-023-12831-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 01/25/2024]
Abstract
The resistance of biofilms to antibiotics is a key factor that makes bacterial infections unsusceptible to antimicrobial therapy. The results of classical tests of cell sensitivity to antibiotics cannot be used to predict therapeutic success in infections associated with biofilm formation. We describe a simple and rapid method for the real-time evaluation of bacterial biofilm sensitivity to antibiotics, with Pseudomonas putida and ampicillin as examples. The method uses an electric biosensor to detect the difference between changes in the biofilm electric polarizability, thereby evaluating antibiotic sensitivity. The electric signals showed that P. putida biofilms were susceptible to ampicillin and that at high antibiotic concentrations, the biofilms differed markedly in their susceptibility (dose-dependent effect). The sensor also detected differences between biofilms before and after ampicillin treatment. The electric-signal changes enabled us to describe the physical picture of the processes occurring in bacterial biofilms in the presence of ampicillin. The approach used in this study is promising for evaluating the activity of various compounds against biofilms, because it permits a conclusion about the antibiotic sensitivity of biofilm bacteria to be made in real time and in a short period (analysis time, not longer than 20 min). An added strong point is that analysis can be done directly in liquid, without preliminary sample preparation. KEY POINTS: • Sensor system to analyze biofilm antimicrobial susceptibility is described. • The signal change depended on the ampicillin concentration (dose-dependent effect). • The sensor allows real-time determination of the antibiofilm effect of ampicillin.
Collapse
Affiliation(s)
- Olga I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms - Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, 410049, Russia.
| | - Stella S Evstigneeva
- Institute of Biochemistry and Physiology of Plants and Microorganisms - Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, 410049, Russia
| | - Alexander A Shirokov
- Institute of Biochemistry and Physiology of Plants and Microorganisms - Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, 410049, Russia
| | | |
Collapse
|
33
|
Elsayed EM, Farghali AA, Zanaty MI, Abdel-Fattah M, Alkhalifah DHM, Hozzein WN, Mahmoud AM. Poly-Gamma-Glutamic Acid Nanopolymer Effect against Bacterial Biofilms: In Vitro and In Vivo Study. Biomedicines 2024; 12:251. [PMID: 38397853 PMCID: PMC10887140 DOI: 10.3390/biomedicines12020251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
In this study, a biodegradable poly-gamma-glutamic-acid nanopolymer (Ɣ-PGA NP) was investigated for its activity against clinical strains of Gram-positive (Staphylococcus aureus and Streptococcus pyogenes) and Gram-negative (Klebsiella pneumoniae and Escherichia coli), and reference strains of S. aureus ATCC 6538, S. pyogenes ATCC 19615 (Gram-positive), and Gram-negative E. coli ATCC 25922, and K. pneumoniae ATCC 13884 bacterial biofilms. The minimum inhibitory concentration (MIC) effect of Ɣ-PGA NP showed inhibitory effects of 0.2, 0.4, 1.6, and 3.2 μg/mL for S. pyogenes, S. aureus, E. coli, and K. pneumoniae, respectively. Also, MIC values were 1.6, 0.8, 0.2, and 0.2 μg/mL for K. pneumoniae ATCC 13884, E. coli ATCC 25922, S. aureus ATCC 6538, and S. pyogenes ATCC 19615, respectively. Afterwards, MBEC (minimum biofilm eradication concentration) and MBIC (minimum biofilm inhibitory concentration) were investigated to detect Ɣ-PGA NPs efficiency against the biofilms. MBEC and MBIC increased with increasing Ɣ-PGA NPs concentration or time of exposure. Interestingly, MBIC values were at lower concentrations of Ɣ-PGA NPs than those of MBEC. Moreover, MBEC values showed that K. pneumoniae was more resistant to Ɣ-PGA NPs than E. coli, S. aureus, and S. pyogenes, and the same pattern was observed in the reference strains. The most effective results for MBEC were after 48 h, which were 1.6, 0.8, 0.4, and 0.2 µg/mL for K. pneumoniae, E. coli, S. aureus, and S. pyogenes, respectively. Moreover, MBIC results were the most impactful after 24 h but some were the same after 48 h. MBIC values after 48 h were 0.2, 0.2, 0.2, and 0.1 μg/mL for K. pneumoniae, E. coli, S. aureus, and S. pyogenes, respectively. The most effective results for MBEC were after 24 h, which were 1.6, 0.8, 0.4, and 0.4 µg/mL for K. pneumoniae ATCC 13884, E. coli ATCC 25922, S. aureus ATCC 6538, and S. pyogenes ATCC 19615, respectively. Also, MBIC results were the most impactful after an exposure time of 12 h. MBIC values after exposure time of 12 h were 0.4, 0.4, 0.2, and 0.2 μg/mL for K. pneumoniae ATCC 13884, E. coli ATCC 25922, S. aureus ATCC 6538, and S. pyogenes ATCC 19615, respectively. Besides that, results were confirmed using confocal laser scanning microscopy (CLSM), which showed a decrease in the number of living cells to 80% and 60% for MBEC and MBIC, respectively, for all the clinical bacterial strains. Moreover, living bacterial cells decreased to 70% at MBEC while decreasing up to 50% at MBIC with all bacterial refence strains. These data justify the CFU quantification. After that, ImageJ software was used to count the attached cells after incubating with the NPs, which proved the variation in live cell count between the manual counting and image analysis methods. Also, a scanning electron microscope (SEM) was used to detect the biofilm architecture after incubation with the Ɣ-PGA NP. In in vivo wound healing experiments, treated wounds of mice showed faster healing (p < 0.00001) than both the untreated mice and those that were only wounded, as the bacterial count was eradicated. Briefly, the infected mice were treated faster (p < 0.0001) when infected with S. pyogenes > S. aureus > E. coli > K. pneumoniae. The same pattern was observed for mice infected with the reference strains. Wound lengths after 2 h showed slightly healing (p < 0.001) for the clinical strains, while treatment became more obvious after 72 h > 48 h > 24 h (p < 0.0001) as wounds began to heal after 24 h up to 72 h. For reference strains, wound lengths after 2 h started to heal up to 72 h.
Collapse
Affiliation(s)
- Eman M. Elsayed
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| | - Ahmed A. Farghali
- Department of Materials Science and Nanotechnology, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Mohamed I. Zanaty
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Medhat Abdel-Fattah
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| | - Dalal Hussien M. Alkhalifah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Wael N. Hozzein
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| | - Ahmed M. Mahmoud
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| |
Collapse
|
34
|
Ghosh S, Sett U, Pal A, Nandy S, Nandi S, Chakrabarty S, Das A, Bandopadhyay P, Basu T. Antibiofilm potential of nanonized eugenol against Pseudomonas aeruginosa. J Appl Microbiol 2024; 135:lxad305. [PMID: 38093454 DOI: 10.1093/jambio/lxad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
AIMS The purpose of this study was to synthesize a nanoform of eugenol (an important phytochemical with various pharmacological potentials) and to investigate its antibiofilm efficacy on Pseudomonas aeruginosa biofilm. METHODS AND RESULTS Colloidal suspension of eugenol-nanoparticles (ENPs) was synthesized by the simple ultrasonic cavitation method through the emulsification of hydrophobic eugenol into hydrophilic gelatin. Thus, the nanonization process made water-insoluble eugenol into water-soluble nano-eugenol, making the nanoform bioavailable. The size of the ENPs was 20-30 nm, entrapment efficiency of eugenol within gelatin was 80%, and release of eugenol from the gelatin cap was slow and sustained over 5 days. Concerning the clinically relevant pathogen P. aeruginosa, ENPs had higher antibiofilm (for both formation and eradication) activities than free eugenol. Minimal biofilm inhibitory concentration and minimal biofilm eradication concentration of ENP on P. aeruginosa biofilm were 2.0 and 4.0 mM, respectively. In addition, the measurement of P. aeruginosa biofilm biomass, biofilm thickness, amount of biofilm extra-polymeric substance, cell surface hydrophobicity, cell swarming and twitching efficiencies, cellular morphology, and biofilm formation in catheter demonstrated that the antibiofilm efficacy of nano-eugenol was 30%-40% higher than that of bulk eugenol. CONCLUSION These results signify that future pharmacological and clinical studies are very much required to investigate whether ENPs can act as an effective drug against P. aeruginosa biofilm-mediated diseases. Thus, the problem of intrinsic antibiotic tolerance of biofilm-forming cells may be minimized by ENPs. Moreover, ENP may be used as a potential catheter-coating agent to inhibit pseudomonal colonization on catheter surfaces and, therefore, to reduce catheter-associated infections and complications.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Upasana Sett
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Anabadya Pal
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Sanchita Nandy
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Susmita Nandi
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Soumajit Chakrabarty
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Abhijit Das
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Pathikrit Bandopadhyay
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Tarakdas Basu
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| |
Collapse
|
35
|
Lunder M, Dahle S, Fink R. Cold atmospheric plasma for surface disinfection: a promising weapon against deleterious meticillin-resistant Staphylococcus aureus biofilms. J Hosp Infect 2024; 143:64-75. [PMID: 37939884 DOI: 10.1016/j.jhin.2023.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/02/2023] [Accepted: 10/15/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Bacteria are becoming increasingly resistant to classical antimicrobial agents, so new approaches need to be explored. AIM To assess the potential of cold atmospheric plasma for the management of meticillin-resistant Staphylococcus aureus (MRSA). METHODS The 24, 48, and 72 h resistant and susceptible S. aureus biofilms were exposed to 60, 120, and 180 s treatment with plasma. FINDINGS Increasing the treatment time results in higher cell reduction for both susceptible and resistant strains of S. aureus (P < 0.05). Up to log10 reduction factor of 5.24 cfu/cm2 can be achieved in 180 s of plasma treatment. Furthermore, plasma can substantially alter the cell's metabolisms and impact cell membrane integrity. However, it has not been shown that plasma can reduce biofilm biomass in the case of 24 h and 48 h biofilms, although the 72 h biofilm was more susceptible, and its biomass was decreased (P < 0.05). The accumulation of intrabacterial reactive oxygen species was also observed, which confirms the plasma's induction of oxidative stress. Finally, it was shown that continuous plasma exposure of bacterial cells does not cause resistance to plasma, nor is resistance developed to cefoxitin. CONCLUSION Cold atmospheric plasma is a good candidate for S. aureus and MRSA biofilm treatment and may therefore be of value in the bacterial resistance crisis.
Collapse
Affiliation(s)
- M Lunder
- University of Ljubljana, Faculty of Health Sciences, Ljubljana, Slovenia
| | - S Dahle
- University of Ljubljana, Biotechnical Faculty, Ljubljana, Slovenia
| | - R Fink
- University of Ljubljana, Faculty of Health Sciences, Ljubljana, Slovenia.
| |
Collapse
|
36
|
Coenye T. Biofilm antimicrobial susceptibility testing: where are we and where could we be going? Clin Microbiol Rev 2023; 36:e0002423. [PMID: 37812003 PMCID: PMC10732061 DOI: 10.1128/cmr.00024-23] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023] Open
Abstract
Our knowledge about the fundamental aspects of biofilm biology, including the mechanisms behind the reduced antimicrobial susceptibility of biofilms, has increased drastically over the last decades. However, this knowledge has so far not been translated into major changes in clinical practice. While the biofilm concept is increasingly on the radar of clinical microbiologists, physicians, and healthcare professionals in general, the standardized tools to study biofilms in the clinical microbiology laboratory are still lacking; one area in which this is particularly obvious is that of antimicrobial susceptibility testing (AST). It is generally accepted that the biofilm lifestyle has a tremendous impact on antibiotic susceptibility, yet AST is typically still carried out with planktonic cells. On top of that, the microenvironment at the site of infection is an important driver for microbial physiology and hence susceptibility; but this is poorly reflected in current AST methods. The goal of this review is to provide an overview of the state of the art concerning biofilm AST and highlight the knowledge gaps in this area. Subsequently, potential ways to improve biofilm-based AST will be discussed. Finally, bottlenecks currently preventing the use of biofilm AST in clinical practice, as well as the steps needed to get past these bottlenecks, will be discussed.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
37
|
Dou J, Ilina P, Cruz CD, Nurmi D, Vidarte PZ, Rissanen M, Tammela P, Vuorinen T. Willow Bark-Derived Material with Antibacterial and Antibiofilm Properties for Potential Wound Dressing Applications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16554-16567. [PMID: 37104679 PMCID: PMC10636761 DOI: 10.1021/acs.jafc.3c00849] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/11/2023]
Abstract
Tree stems contain wood in addition to 10-20% bark, which remains one of the largest underutilized biomasses on earth. Unique macromolecules (like lignin, suberin, pectin, and tannin), extractives, and sclerenchyma fibers form the main part of the bark. Here, we perform detailed investigation of antibacterial and antibiofilm properties of bark-derived fiber bundles and discuss their potential application as wound dressing for treatment of infected chronic wounds. We show that the yarns containing at least 50% of willow bark fiber bundles significantly inhibit biofilm formation by wound-isolated Staphylococcus aureus strains. We then correlate antibacterial effects of the material to its chemical composition. Lignin plays the major role in antibacterial activity against planktonic bacteria [i.e., minimum inhibitory concentration (MIC) 1.25 mg/mL]. Acetone extract (unsaturated fatty acid-enriched) and tannin-like (dicarboxylic acid-enriched) substances inhibit both bacterial planktonic growth [MIC 1 and 3 mg/mL, respectively] and biofilm formation. The yarn lost its antibacterial activity once its surface lignin reached 20.1%, based on X-ray photoelectron spectroscopy. The proportion of fiber bundles at the fabricated yarn correlates positively with its surface lignin. Overall, this study paves the way to the use of bark-derived fiber bundles as a natural-based material for active (antibacterial and antibiofilm) wound dressings, upgrading this underappreciated bark residue from an energy source into high-value pharmaceutical use.
Collapse
Affiliation(s)
- Jinze Dou
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, 00076 Aalto, Finland
| | - Polina Ilina
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Cristina D. Cruz
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Denise Nurmi
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, 00076 Aalto, Finland
| | - Paula Zegarra Vidarte
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Marja Rissanen
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, 00076 Aalto, Finland
| | - Päivi Tammela
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Tapani Vuorinen
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, 00076 Aalto, Finland
| |
Collapse
|
38
|
Pourhajibagher M, Azimi Mohammadabadi M, Ghafari HA, Hodjat M, Bahador A. Evaluation of anti-biofilm effect of antimicrobial sonodynamic therapy-based periodontal ligament stem cell-derived exosome-loaded kojic acid on Enterococcus faecalis biofilm. J Med Microbiol 2023; 72. [PMID: 37910015 DOI: 10.1099/jmm.0.001772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Introduction. Antimicrobial sonodynamic therapy (aSDT) is an approach that uses ultrasound waves (UWs) and a sonosensitizer to generate reactive oxygen species (ROS) to damage microbial cells in biofilms. Using nano-carriers, such as exosomes (Exos), to deliver the sonosensitizer can potentially enhance the effectiveness of aSDT.Hypothesis/Gap Statement. aSDT can downregulate the expression of gelE and sprE genes, increasing the production of endogenous ROS and degradation of pre-formed Enterococcus faecalis biofilms.Aim. This study investigated the anti-biofilm effect of aSDT-based periodontal ligament stem cell-derived exosome-loaded kojic acid (KA@PDL-Exo) on pre-formed E. faecalis biofilms in root canals.Methodology. Following the isolation and characterization of PDL-Exo, KA@PDL-Exo was prepared and confirmed. The minimal biofilm inhibitory concentration (MBIC) of KA, PDL-Exo, KA@PDL-Exo and sodium hypochlorite (NaOCl) was determined, and their anti-biofilm effects were assessed with and without UWs. The binding affinity of KA with GelE and SprE proteins was evaluated using in silico molecular docking. Additionally, the study measured the generation of endogenous ROS and evaluated changes in the gene expression levels of gelE and sprE.Results. The results revealed a dose-dependent decrease in the viability of E. faecalis cells within biofilms. KA@PDL-Exo was the most effective, with an MBIC of 62.5 µg ml-1, while NaOCl, KA and PDL-Exo had MBIC values of 125, 250 and 500 µg ml-1, respectively. The use of KA@PDL-Exo-mediated aSDT resulted in a significant reduction of the E. faecalis biofilm (3.22±0.36 log10 c.f.u. ml-1; P<0.05). The molecular docking analysis revealed docking scores of -5.3 and -5.2 kcal mol-1 for GelE-KA an SprE-KA, respectively. The findings observed the most significant reduction in gene expression of gelE and sprE in the KA@PDL-Exo group, with a decrease of 7.9- and 9.3-fold, respectively, compared to the control group (P<0.05).Conclusion. The KA@PDL-Exo-mediated aSDT was able to significantly reduce the E. faecalis load in pre-formed biofilms, decrease the expression of gelE and srpE mRNA, and increase the generation of endogenous ROS. These findings imply that KA@PDL-Exo-mediated aSDT could be a promising anti-biofilm strategy that requires additional in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Azimi Mohammadabadi
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hassan-Ali Ghafari
- Department of Orthodontics, School of Dentistry, Shahed University, Tehran, Iran
| | - Mahshid Hodjat
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Bahador
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Fellowship in Clinical Laboratory Sciences, BioHealth Lab, Tehran, Iran
| |
Collapse
|
39
|
Carmona-Orozco ML, Quiñones W, Robledo SM, Torres F, Echeverri F. Reversing the biofilm-inducing effect of two xanthones from Garcinia mangostana by 3-methyl-2(5H)-furanone and N-butyryl-D-L homoserine lactone. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155069. [PMID: 37722186 DOI: 10.1016/j.phymed.2023.155069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/26/2023] [Accepted: 09/04/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND According to the WHO, 12 bacteria cause numerous human infections, including Enterobacteriaceae Klebsiella pneumoniae, and thus represent a public health problem. Microbial resistance is associated with biofilm formation; therefore, it is critical to know the biofilm-inducing potential of various compounds of everyday life. Likewise, the reversibility of biofilms and the modulation of persister cells are important for controlling microbial pathogens. In this work, we investigated the biofilm-inducing effects of xanthones from Garcinia mangostana on Klebsiella pneumoniae. Furthermore, we investigated the reversal effect of 3-methyl-2(5H)-furanone and the formation of persister cells induced by xanthones and their role in modulating the biofilm to the antibiotic gentamicin. METHODS To analyze the biofilm-inducing role of xanthones from Garcinia mangostana, cultures of K. pneumoniae containing duodenal probe pieces were treated with 0.1-0.001 μM α- and γ-mangostin, and the biofilm levels were measured using spectrophotometry. To determine biofilm reversion, cultures treated with xanthones, or gentamicin were mixed with 3-methyl-2(5H)-furanone or N-butyryl-DL-homoserine lactone. The presence of K. pneumoniae persister cells was determined by applying the compounds to the mature biofilm, and the number of colony-forming units was counted. RESULTS The xanthones α- and γ-mangostin increased K. pneumoniae biofilm production by 40% with duodenal probes. However, 3-methyl-2(5H)-furanone at 0.001 μΜ reversed biofilm formation by up to 60%. Moreover, adding the same to a culture treated with gentamicin reduced the biofilm by 80.5%. This effect was highlighted when 3-methyl-2(5H)-furanone was administered 6 h later than xanthones. At high concentrations of α-mangostin, persister K. pneumoniae cells in the biofilm were about 5 - 10 times more abundant than cells, whereas, with γ-mangostin, they were about 100 times more. CONCLUSION Two xanthones, α- and γ-mangostin from G. mangostana, induced biofilm formation in K. pneumoniae and promoted persister cells. However, the biofilm formation was reversed by adding 3-methyl-2(5H)-furanone, and even this effect was achieved with gentamicin. In addition, this compound controlled the persister K. pneumoniae cells promoted by α-mangostin. Thus, synthetic, and natural biofilm-inducing compounds could harm human health. Therefore, avoiding these substances and looking for biofilm inhibitors would be a strategy to overcome microbial resistance and recover antibiotics that are no longer used.
Collapse
Affiliation(s)
- Maria L Carmona-Orozco
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia
| | - Wiston Quiñones
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia
| | - Sara M Robledo
- PECET-Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Fernando Torres
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia
| | - Fernando Echeverri
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
40
|
Selmi H, Rocchetti MT, Capozzi V, Semedo-Lemsaddek T, Fiocco D, Spano G, Abidi F. Lactiplantibacillus plantarum from Unexplored Tunisian Ecological Niches: Antimicrobial Potential, Probiotic and Food Applications. Microorganisms 2023; 11:2679. [PMID: 38004691 PMCID: PMC10673251 DOI: 10.3390/microorganisms11112679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The continued exploration of the diversity of lactic acid bacteria in little-studied ecological niches represents a fundamental activity to understand the diffusion and biotechnological significance of this heterogeneous class of prokaryotes. In this study, Lactiplantibacillus plantarum (Lpb. plantarum) strains were isolated from Tunisian vegetable sources, including fermented olive and fermented pepper, and from dead locust intestines, which were subsequently evaluated for their antimicrobial activity against foodborne pathogenic bacteria, including Escherichia coli O157:H7 CECT 4267 and Listeria monocytogenes CECT 4031, as well as against some fungi, including Penicillium expansum, Aspergilus niger, and Botrytis cinerea. In addition, their resistance to oro-gastro-intestinal transit, aggregation capabilities, biofilm production capacity, adhesion to human enterocyte-like cells, and cytotoxicity to colorectal adenocarcinoma cell line were determined. Further, adhesion to tomatoes and the biocontrol potential of this model food matrix were analyzed. It was found that all the strains were able to inhibit the indicator growth, mostly through organic acid production. Furthermore, these strains showed promising probiotic traits, including in vitro tolerance to oro-gastrointestinal conditions, and adhesion to abiotic surfaces and Caco-2 cells. Moreover, all tested Lpb. plantarum strains were able to adhere to tomatoes with similar rates (4.0-6.0 LogCFU/g tomato). The co-culture of LAB strains with pathogens on tomatoes showed that Lpb. plantarum could be a good candidate to control pathogen growth. Nonetheless, further studies are needed to guarantee their use as probiotic strains for biocontrol on food matrices.
Collapse
Affiliation(s)
- Hiba Selmi
- Faculty of Sciences of Bizerte, University of Carthage, Zarzouna, Bizerte 7021, Tunisia
- Laboratory of Protein Engineering and Bioactive Molecules (LIP-MB), National Institute of Applied Sciences and Technology, University of Carthage, Carthage 1054, Tunisia;
| | - Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| | - Vittorio Capozzi
- Institute of Sciences of Food Production, National Research Council (CNR) of Italy, c/o CS-DAT, Via Michele Protano, 71122 Foggia, Italy;
| | - Teresa Semedo-Lemsaddek
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Av. da Universidade Técnica de Lisboa, 1300-477 Lisbon, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| | - Giuseppe Spano
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy
| | - Ferid Abidi
- Laboratory of Protein Engineering and Bioactive Molecules (LIP-MB), National Institute of Applied Sciences and Technology, University of Carthage, Carthage 1054, Tunisia;
| |
Collapse
|
41
|
Liu T, Zhai Y, Jeong KC. Advancing understanding of microbial biofilms through machine learning-powered studies. Food Sci Biotechnol 2023; 32:1653-1664. [PMID: 37780593 PMCID: PMC10533454 DOI: 10.1007/s10068-023-01415-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 10/03/2023] Open
Abstract
Microbial biofilms are prevalent in various environments and pose significant challenges to food safety and public health. The biofilms formed by pathogens can cause food spoilage, foodborne illness, and infectious diseases, which are difficult to treat due to their enhanced antimicrobial resistance. While the composition and development of biofilms have been widely studied, their profound impact on food, the food industry, and public health has not been sufficiently recapitulated. This review aims to provide a comprehensive overview of microbial biofilms in the food industry and their implication on public health. It highlights the existence of biofilms along the food-producing chains and the underlying mechanisms of biofilm-associated diseases. Furthermore, this review thoroughly summarizes the enhanced understanding of microbial biofilms achieved through machine learning approaches in biofilm research. By consolidating existing knowledge, this review intends to facilitate developing effective strategies to combat biofilm-associated infections in both the food industry and public health.
Collapse
Affiliation(s)
- Ting Liu
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32610 USA
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32608 USA
| | - Yuting Zhai
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32610 USA
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32608 USA
| | - Kwangcheol Casey Jeong
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32610 USA
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32608 USA
| |
Collapse
|
42
|
Kim SH, Lee H, Park MK. Isolation, characterization, and application of a novel, lytic phage vB_SalA_KFSST3 with depolymerase for the control of Salmonella and its biofilm on cantaloupe under cold temperature. Food Res Int 2023; 172:113062. [PMID: 37689855 DOI: 10.1016/j.foodres.2023.113062] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 09/11/2023]
Abstract
This study investigated the efficacy of a novel Salmonella phage with depolymerase activity to control S. Typhimurium (ST) and its biofilm on cantaloupes, for the first time, under simulated cold temperature. vB_SalA_KFSST3 forming a halo zone was isolated and purified from a slaughterhouse with a final concentration of 12.1 ± 0.1 log PFU/mL. Based on the morphological and bioinformatics analyses, vB_SalA_KFSST3 was identified as a novel phage belonging to the family Ackermannviridae. Before employing the phage on cantaloupe, its genetic characteristics, specificity, stability, and bactericidal effect were investigated. Genetic analyses confirmed its safety and identified endolysin and two depolymerase domains possessing antibiofilm potential. In addition, the phage exhibited a broad specificity with great efficiencies toward five Salmonella strains at 4 °C, 22 °C, and 37 °C, as well as stable lytic activity over a wide range of pHs (3 to 11) and temperatures (-20 °C to 60 °C). The optimal multiplicity of infection (MOI) and exposure time of phage were determined to be 100 and 2 h, respectively, based on the highest bacterial reduction of ∼2.7 log CFU/mL. Following the formation of ST biofilm on cantaloupe at 4 °C and 22 °C, the cantaloupe was treated with phage at an MOI of 100 for 2 h. The antibiofilm efficacy of phage was evaluated via the plate count method, confocal laser scanning microscopy, and scanning electron microscopy (SEM). The initial biofilm population at 22 °C was significantly greater and more condensed than that at 4 °C. After phage treatment, biofilm population and the percentage of viable ST in biofilm were reduced by ∼4.6 log CFU/cm2 and ∼90% within 2 h, respectively, which were significantly greater than those at 22 °C (∼2.0 log CFU/cm2 and ∼45%) (P < 0.05). SEM images also confirmed more drastic destruction of the cohesive biofilm architecture at 4 °C than at 22 °C. As a result of its cold temperature-robust lytic activity and the contribution of endolysin and two depolymerases, vB_SalA_KFSST3 demonstrated excellent antibiofilm efficacy at cold temperature, highlighting its potential as a promising practical biocontrol agent for the control of ST and its biofilm.
Collapse
Affiliation(s)
- Su-Hyeon Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Heejeong Lee
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mi-Kyung Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea; Food and Bio-Industry Institute, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
43
|
Wei M, Wang P, Li T, Wang Q, Su M, Gu L, Wang S. Antimicrobial and antibiofilm effects of essential fatty acids against clinically isolated vancomycin-resistant Enterococcus faecium. Front Cell Infect Microbiol 2023; 13:1266674. [PMID: 37842001 PMCID: PMC10570806 DOI: 10.3389/fcimb.2023.1266674] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Enterococcus faecium is a leading cause of hospital-acquired infections, which has become a serious public health concern. The increasing incidence of vancomycin-resistant E. faecium (VRE-fm) raises an urgent need to find new antimicrobial agents as a complement to traditional antibiotics. The study aimed to evaluate the antimicrobial and antibiofilm activity of essential fatty acids (EFAs) against VRE-fm, and further explore the molecular mechanism of the antibiofilm activity of EFAs. Method The microdilution broth method was used for antimicrobial susceptibility testing with traditional antibiotics and EFAs, including α-linolenic acid (ALA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), linoleic acid (LOA), γ-linolenic acid (GLA), and arachidonic acid (AA). The effect of EFAs on cell morphology of VRE-fm was investigated by scanning electron microscopy. The crystal violet method was used to evaluate the antibiofilm activities of EFAs against VRE-fm. Furthermore, the expression of biofilm-related genes (acm, atlA, esp, and sagA) of VRE-fm isolates under the action of GLA was analyzed using quantitative reverse transcription PCR (qRT-PCR) assay. Results VRE-fm isolates were highly resistant to most traditional antibiotics, only highly susceptible to quinupristin-dalfopristin (90.0%), tigecycline (100%), and linezolid (100%). EPA, DHA, and GLA exhibited excellent antimicrobial activity. The MIC50/90 of EPA, DHA, and GLA were 0.5/1, 0.25/0.5, and 0.5/1 mM, respectively. SEM imaging showed that strain V27 adsorbed a large number of DHA molecules. Furthermore, all EFAs exhibited excellent inhibition and eradication activities against VRE-fm biofilms. The biofilm inhibition rates of EFAs ranged from 45.3% to 58.0%, and eradication rates ranged from 54.1% to 63.4%, against 6 VRE-fm isolates with moderate biofilm formation ability. GLA exhibited remarkable antibiofilm activity against VRE-fm isolates. The qRT-PCR analysis showed that GLA could significantly down-regulate the expression of the atlA gene (P < 0.01) of VRE-fm. Conclusion DHA showed the strongest antibacterial activity, while GLA showed the strongest antibiofilm effect among the EFAs with antibacterial activity. Our novel findings indicate that the antibiofilm activity of GLA may be through down-regulating the atlA gene expression in VRE-fm. Therefore, DHA and GLA had the potential to be developed as therapeutic agents to treat infections related to multiple antimicrobial-resistant E. faecium.
Collapse
Affiliation(s)
- Ming Wei
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Peng Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Tianmeng Li
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qiangyi Wang
- Department of Clinical Laboratory, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingze Su
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Li Gu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuai Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Enriquez KT, Plummer WD, Neufer PD, Chazin WJ, Dupont WD, Skaar EP. Temporal modelling of the biofilm lifecycle (TMBL) establishes kinetic analysis of plate-based bacterial biofilm dynamics. J Microbiol Methods 2023; 212:106808. [PMID: 37595876 PMCID: PMC10528067 DOI: 10.1016/j.mimet.2023.106808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/12/2023] [Accepted: 08/12/2023] [Indexed: 08/20/2023]
Abstract
Bacterial biofilms are critical to pathogenesis and infection. They are associated with rising rates of antimicrobial resistance. Biofilms are correlated with worse clinical outcomes, making them important to infectious diseases research. There is a gap in knowledge surrounding biofilm kinetics and dynamics which makes biofilm research difficult to translate from bench to bedside. To address this gap, this work employs a well-characterized crystal violet biomass accrual and planktonic cell density assay across a clinically relevant time course and expands statistical analysis to include kinetic information in a protocol termed the TMBL (Temporal Mapping of the Biofilm Lifecycle) assay. TMBL's statistical framework quantitatively compares biofilm communities across time, species, and media conditions in a 96-well format. Measurements from TMBL can reliably be condensed into response features that inform the time-dependent behavior of adherent biomass and planktonic cell populations. Staphylococcus aureus and Pseudomonas aeruginosa biofilms were grown in conditions of metal starvation in nutrient-variable media to demonstrate the rigor and translational potential of this strategy. Significant differences in single-species biofilm formation are seen in metal-deplete conditions as compared to their controls which is consistent with the consensus literature on nutritional immunity that metal availability drives transcriptomic and metabolomic changes in numerous pathogens. Taken together, these results suggest that kinetic analysis of biofilm by TMBL represents a statistically and biologically rigorous approach to studying the biofilm lifecycle as a time-dependent process. In addition to current methods to study the impact of microbe and environmental factors on the biofilm lifecycle, this kinetic assay can inform biological discovery in biofilm formation and maintenance.
Collapse
Affiliation(s)
- Kyle T Enriquez
- Vanderbilt University Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, United States of America; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States of America; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - W Dale Plummer
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States of America
| | - Preston D Neufer
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America; Department of Biochemistry, Vanderbilt University, Nashville, TN, United States of America
| | - Walter J Chazin
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States of America; Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America; Department of Biochemistry, Vanderbilt University, Nashville, TN, United States of America
| | - William D Dupont
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States of America
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States of America; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States of America.
| |
Collapse
|
45
|
Mishra RAK, Muthukaliannan GK, Rathinasabapathi P. Effects of Flavonoids and Antibiotics Combination on Preformed Biofilms and Small RNA of Staphylococcus aureus. Indian J Microbiol 2023; 63:307-316. [PMID: 37781018 PMCID: PMC10533456 DOI: 10.1007/s12088-023-01086-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 07/24/2023] [Indexed: 10/03/2023] Open
Abstract
Antibiotic resistance of Staphylococcus aureus has considerably increased among non-clinical or asymptomatic individuals. The formation of biofilms denies antimicrobial access to its targets present on the surface and inside the cell. The present study tested the effect of the combination of flavonoids and antibiotics over the preformed biofilms of S. aureus. The eradication of the preformed biofilms was analyzed using the crystal violet method. It has shown that 2500 µg mL-1 Rutin and 100 µg mL-1 Erythromycin (MIC Concentration) combination efficiently reduced the growth of the cells, which were adhered to the surfaces forming the biofilms. Fluorescence microscopic analysis indicated that the Rutin and Erythromycin (MIC value) combinations could eradicate the preformed biofilm cells more efficiently than other combinations. We found that the flavonoids and antibiotics with MIC concentration show a significant effect over the preformed biofilms cells of S. aureus. In addition, the semi-quantitative real-time PCR analysis for the sRNAs under the treatment of Rutin and Erythromycin combinations showed that few small RNAs expression (SprF, SprG, ArtR, Teg49, Teg41, and RNAIII) are getting downregulated upon the treatment; but again recovers with the incubation time interval increases. Combinations have a significant effect on Teg49 where there is a very faint intensity of the band, but for other small RNAs, there is an irregular pattern on the gel image. It has been concluded that at the initial period of incubation, the combinations have an effect on all the sRNAs but once the incubation increases, the effects have been slowly decreasing. It has been concluded that the combination has been able to reduce the doubling time of S. aureus upon treatment. Whereas, the small RNAs used in the study can be further evaluated for expression profiling through qRT-PCT. Graphical abstract
Collapse
Affiliation(s)
- Rudra Awdhesh Kumar Mishra
- School of Biosciences and Technology, Vellore Institute of Technology, Katpadi, Vellore, 632014 India
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, 603203 Tamil Nadu India
| | | | - Pasupathi Rathinasabapathi
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, 603203 Tamil Nadu India
| |
Collapse
|
46
|
Ray S, Jin JO, Choi I, Kim M. Cell-Free Supernatant of Bacillus thuringiensis Displays Anti-Biofilm Activity Against Staphylococcus aureus. Appl Biochem Biotechnol 2023; 195:5379-5393. [PMID: 35593953 DOI: 10.1007/s12010-022-03971-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/10/2022] [Indexed: 11/02/2022]
Abstract
Staphylococcus aureus is an important bacterial pathogen responsible for biofilm formation in medical devices. Due to the increasing antibiotic resistance of S. aureus, it is necessary to search for new anti-biofilm agents. In this study, the cell-free supernatant of Bacillus thuringiensis inhibited biofilm formation up to 93% and dispersed biofilms up to 83% without affecting the growth of S. aureus. The ethyl acetate extract of B. thuringiensis cell-free supernatant exhibited a dose-dependent anti-biofilm activity against S. aureus with the biofilm inhibition concentration ranging from 8 to 64 µg/mL. Scanning electron microscopy revealed that the cell-free supernatant extract of B. thuringiensis resulted in a significant reduction in S. aureus biofilms. The ethyl acetate extract of cell-free supernatant of B. thuringiensis was found to contain various compounds with structural similarity to known anti-biofilm compounds. In particular, squalene, cinnamic acid derivatives, and eicosapentaene seem to act synergistically against S. aureus biofilms. Hence, B. thuringiensis cell-free supernatant proved to be effective against S. aureus biofilms. The results clearly show the potential of natural molecules produced by B. thuringiensis as alternative therapies with anti-biofilm activity instead of bactericidal properties.
Collapse
Affiliation(s)
- Subhasree Ray
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Jun-O Jin
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea
| | - Inho Choi
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea
| | - Myunghee Kim
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea.
- Department of Food Science and Technology, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea.
| |
Collapse
|
47
|
Chen W, Xu Z, Li C, Wang C, Wang M, Liang J, Wei P. Investigation of biofilm formation and the associated genes in multidrug-resistant Salmonella pullorum in China (2018-2022). Front Vet Sci 2023; 10:1248584. [PMID: 37720478 PMCID: PMC10500350 DOI: 10.3389/fvets.2023.1248584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023] Open
Abstract
The study explored the biofilm (BF) formation capacity, BF-related gene profiles, and the trends in antimicrobial resistance (AMR) of Salmonella pullorum (SP) strains over several years. A total of 627 SP strains were isolated from 4,540 samples collected from chicken farms in Guangxi, China during 2018-2022. The BF-forming capacity of these isolates was assessed using crystal violet staining, and the presence of eight BF-related genes (csgA, csgB, csgD, ompR, bapA, pfs, luxS, and rpoS) in BF formation-positive strains was determined through Polymerase Chain Reaction (PCR) analysis. Antimicrobial susceptibility test was conducted to investigate the AMR of the isolates. Minimum Inhibitory Concentration (MIC) and Minimal Biofilm Eradication Concentration (MBEC) of nine SP-BF strains were determined using the broth microdilution method to assess the impact of BF formation on AMR. Additionally, the Optimal Biofilm Formation Conditions (OBFC) were investigated. The results indicated that 36.8% (231/627) of the strains exhibited a positive BF-formation capacity. Among these, 24.7% (57/231) were strong BF producers, 23.4% (54/231) were moderate BF producers, and 51.9% (120/231) were weak BF producers. Analysis of the eight BF-related genes in SP-BF strains revealed that over 90% of them were positive for all the genes. Antimicrobial susceptibility test conducted on the isolates showed that 100% (231/231) of them exhibited resistance to at least one antibiotic, with 98.3% (227/231) demonstrating multidrug resistance (MDR). Both MIC and MBEC measurements indicated varying degrees of increased AMR after BF formation of the bacteria. The optimal conditions for BF formation were observed at 37°C after 48 h of incubation, with an initial bacterial concentration of 1.2 × 106 CFU/mL. Notably, NaCl had a significant inhibitory effect on BF formation, while glucose and Trypticase Soy Broth (TSB) positively influenced BF formation. The results of the study emphasized the need for effective preventive and control strategies to address the challenges posed by the BF formation and MDR of SP in the field.
Collapse
Affiliation(s)
- Wenyan Chen
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| | - Ziheng Xu
- School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Changcheng Li
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| | - Can Wang
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| | - Min Wang
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| | - Jingzhen Liang
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| | - Ping Wei
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| |
Collapse
|
48
|
Samaniego LVB, Higasi PMR, de Mello Capetti CC, Cortez AA, Pratavieira S, de Oliveira Arnoldi Pellegrini V, Dabul ANG, Segato F, Polikarpov I. Staphylococcus aureus microbial biofilms degradation using cellobiose dehydrogenase from Thermothelomyces thermophilus M77. Int J Biol Macromol 2023; 247:125822. [PMID: 37451383 DOI: 10.1016/j.ijbiomac.2023.125822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
This work reports biochemical characterization of Thermothelomyces thermophilus cellobiose dehydrogenase (TthCDHIIa) and its application as an antimicrobial and antibiofilm agent. We demonstrate that TthCDHIIa is thermostable in different ionic solutions and is capable of oxidizing multiple mono and oligosaccharide substrates and to continuously produce H2O2. Kinetics measurements depict the enzyme catalytic characteristics consistent with an Ascomycota class II CDH. Our structural analyses show that TthCDHIIa substrate binding pocket is spacious enough to accommodate larger cello and xylooligosaccharides. We also reveal that TthCDHIIa supplemented with cellobiose reduces the viability of S. aureus ATCC 25923 up to 32 % in a planktonic growth model and also inhibits its biofilm growth on 62.5 %. Furthermore, TthCDHIIa eradicates preformed S. aureus biofilms via H2O2 oxidative degradation of the biofilm matrix, making these bacteria considerably more susceptible to gentamicin and tetracycline.
Collapse
Affiliation(s)
| | - Paula Miwa Rabelo Higasi
- Sao Carlos Institute of Physics, University of Sao Paulo, 1100 João Dagnone Avenue, 13563-120 São Carlos, SP, Brazil
| | - Caio Cesar de Mello Capetti
- Sao Carlos Institute of Physics, University of Sao Paulo, 1100 João Dagnone Avenue, 13563-120 São Carlos, SP, Brazil
| | - Anelyse Abreu Cortez
- Sao Carlos Institute of Physics, University of Sao Paulo, 1100 João Dagnone Avenue, 13563-120 São Carlos, SP, Brazil
| | - Sebastião Pratavieira
- Sao Carlos Institute of Physics, University of Sao Paulo, 1100 João Dagnone Avenue, 13563-120 São Carlos, SP, Brazil
| | | | - Andrei Nicoli Gebieluca Dabul
- Sao Carlos Institute of Physics, University of Sao Paulo, 1100 João Dagnone Avenue, 13563-120 São Carlos, SP, Brazil
| | - Fernando Segato
- Lorena School of Engineering, University of Sao Paulo, Estrada Municipal do Campinho, 12602-810 Lorena, SP, Brazil
| | - Igor Polikarpov
- Sao Carlos Institute of Physics, University of Sao Paulo, 1100 João Dagnone Avenue, 13563-120 São Carlos, SP, Brazil.
| |
Collapse
|
49
|
Pangprasit N, Thammawong Y, Kulsirorat A, Chuammitri P, Kongkaew A, Intanon M, Suriyasathaporn W, Pikulkaew S, Chaisri W. Titanium Dioxide Nano-Formulation: Characterization, Antimicrobial Activity, and Wound Healing in Animals. Animals (Basel) 2023; 13:2688. [PMID: 37684952 PMCID: PMC10486583 DOI: 10.3390/ani13172688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/04/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
The use of metal oxide nanoparticles as an alternative antimicrobial agent has gained attention due to the increasing problem of antimicrobial resistance. Understanding its properties and potential benefits can contribute to the development of more effective and sustainable treatments in veterinary medicine. The aim of this study was to characterize TiO2-NP formulations and evaluate their antibacterial and wound healing abilities. The diameters and zeta potentials were determined using the Zetasizer in conjunction with dynamic light scattering. The agar-well diffusion method, time-kill kinetic assay and crystal violet assay were used to evaluate their antimicrobial activities. Wound healing assays were conducted both in vitro and in vivo. The study demonstrated that TiO2-NP formulations exhibit significant antimicrobial properties against various bacterial strains such as S. aureus and E. coli. No measurable E. coli growth was observed within a 15-min period following exposure to TiO2-NP formulations. The TiO2-NP formation can improve wound healing by enhancing cell migration and collagen formation in both in vitro and in vivo conditions. In summary, our study suggests that TiO2-NP has the potential for use as an antimicrobial agent for animal wound treatment due to its ability to suppress bacterial growth and biofilm formation, as well as to enhance wound healing.
Collapse
Affiliation(s)
- Noppason Pangprasit
- PhD’s Degree Program, Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (Y.T.); (A.K.); (W.S.); (S.P.)
| | - Yada Thammawong
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (Y.T.); (A.K.); (W.S.); (S.P.)
| | - Alongkorn Kulsirorat
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (Y.T.); (A.K.); (W.S.); (S.P.)
| | - Phongsakorn Chuammitri
- Department of Veterinary Bioscience and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (P.C.); (M.I.)
- Research Center of Producing and Development of Products and Innovations for Animal Health, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Montira Intanon
- Department of Veterinary Bioscience and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (P.C.); (M.I.)
- Research Center of Producing and Development of Products and Innovations for Animal Health, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Witaya Suriyasathaporn
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (Y.T.); (A.K.); (W.S.); (S.P.)
- Research Center of Producing and Development of Products and Innovations for Animal Health, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Surachai Pikulkaew
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (Y.T.); (A.K.); (W.S.); (S.P.)
- Research Center of Producing and Development of Products and Innovations for Animal Health, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Wasana Chaisri
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (Y.T.); (A.K.); (W.S.); (S.P.)
- Research Center of Producing and Development of Products and Innovations for Animal Health, Chiang Mai University, Chiang Mai 50100, Thailand
| |
Collapse
|
50
|
Tran NN, Morrisette T, Jorgensen SCJ, Orench-Benvenutti JM, Kebriaei R. Current therapies and challenges for the treatment of Staphylococcus aureus biofilm-related infections. Pharmacotherapy 2023; 43:816-832. [PMID: 37133439 DOI: 10.1002/phar.2806] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 05/04/2023]
Abstract
Staphylococcus aureus is a major cause of nosocomial and community-acquired infections and contributes to significant increase in morbidity and mortality especially when associated with medical devices and in biofilm form. Biofilm structure provides a pathway for the enrichment of resistant and persistent phenotypes of S. aureus leading to relapse and recurrence of infection. Minimal diffusion of antibiotics inside biofilm structure leads to heterogeneity and distinct physiological activity. Additionally, horizontal gene transfer between cells in proximity adds to the challenges associated with eradication of biofilms. This narrative review focuses on biofilm-associated infections caused by S. aureus, the impact of environmental conditions on biofilm formation, interactions inside biofilm communities, and the clinical challenges that they present. Conclusively, potential solutions, novel treatment strategies, combination therapies, and reported alternatives are discussed.
Collapse
Affiliation(s)
- Nikki N Tran
- Department of Pharmacy, The Ohio State University Wexner Medical Center - The James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | - Taylor Morrisette
- Department of Clinical Pharmacy and Outcomes Sciences, Medical University of South Carolina College of Pharmacy, Charleston, South Carolina, USA
- Department of Pharmacy Services, Medical University of South Carolina Shawn Jenkins Children's Hospital, Charleston, South Carolina, USA
| | - Sarah C J Jorgensen
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - José M Orench-Benvenutti
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Razieh Kebriaei
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|