1
|
Kranjec C, Mathew JP, Ovchinnikov K, Fadayomi I, Yang Y, Kjos M, Li WW. A bacteriocin-based coating strategy to prevent vancomycin-resistant Enterococcus faecium biofilm formation on materials of interest for indwelling medical devices. Biofilm 2024; 8:100211. [PMID: 39071174 PMCID: PMC11282937 DOI: 10.1016/j.bioflm.2024.100211] [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: 11/27/2023] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
The ever-increasing use of exogenous materials as indwelling medical devices in modern medicine offers to pathogens new ways to gain access to human body and begin, in some cases, life threatening infections. Biofouling of such materials with bacteria or fungi is a major concern during surgeries, since this is often associated with biofilm formation and difficult to treat, recalcitrant infections. Intense research efforts have therefore developed several strategies to shield the medical devices' surface from colonization by pathogenic microorganisms. Here, we used dopamine as a coupling agent to coat four different materials of medical interest (plastic polyetheretherketone (PEEK), stainless steel, titanium and silicone catheter) with the bacteriocins, enterocin EJ97-short and the thiopeptide micrococcin P1. Water contact angle measurements and x-ray photoelectron spectroscopy were used to verify the effective coating of the materials. The effect of bacteriocins coated on these materials on the biofilm formation by a vancomycin resistant Enterococcus faecium (VRE) strain was studied by biofilm-oriented antimicrobial test (BOAT) and electron scanning microscopy. The in vitro biocompatibility of bacteriocin-modified biomaterials was tested on cultured human cells. The results demonstrated that the binding of the bacteriocins to the implant surfaces is achieved, and the two bacteriocins in combination could inhibit biofilm formation by E. faecium on all four materials. The modified implant showed no cytotoxicity to the human cells tested. Therefore, surface modification with the two bacteriocins may offer a novel and effective way to prevent biofilm formation on a wide range of implant materials.
Collapse
Affiliation(s)
- Christian Kranjec
- Laboratory of Microbial Gene Technology, Faculty of Chemistry, Biotechnology and Food Science. Norwegian University of Life Sciences, 1430, Ås, Norway
| | - Jills Puthiaparambil Mathew
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Kirill Ovchinnikov
- Laboratory of Microbial Gene Technology, Faculty of Chemistry, Biotechnology and Food Science. Norwegian University of Life Sciences, 1430, Ås, Norway
| | - Idowu Fadayomi
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Ying Yang
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Morten Kjos
- Laboratory of Microbial Gene Technology, Faculty of Chemistry, Biotechnology and Food Science. Norwegian University of Life Sciences, 1430, Ås, Norway
| | - Wen-Wu Li
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, ST4 7QB, UK
| |
Collapse
|
2
|
Vadakkan K, Hemapriya J, Ngangbam AK, Sathishkumar K, Mapranathukaran VO. Biofilm inhibition of Staphylococcus aureus by silver nanoparticles derived from Hellenia speciosa rhizome extract. Microb Pathog 2024; 196:106933. [PMID: 39270757 DOI: 10.1016/j.micpath.2024.106933] [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: 05/28/2024] [Revised: 08/02/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Staphylococcus aureus is the most common cause of serious health conditions because of the formation of biofilm, which lowers antibiotic efficacy and enhances infection transmission and tenacious behavior. This bacteria is a major threat to the worldwide healthcare system. Silver nanoparticles have strong antibacterial characteristics and emerged as a possible alternative. This work is most relevant since it investigates the parameters influencing the biogenic nanoparticle-assisted control of bacterial biofilms by Staphylococcus aureus. Nanoparticles were fabricated utilizing Hellenia speciosa rhizome extracts, which largely comprised physiologically active components such as spirost-5-en-3-yl acetate, thymol, stigmasterol, and diosgenin, enhanced with the creation of silver nanocomposites. GC-MS, XRD, DLS, SEM, EDX, FTIR and TEM were used to investigate the characteristics of nanoparticles. The microtiter plate experiment showed that nanoparticles destroyed biofilms by up to 92.41 % at doses that ranged from 0 to 25 μg/ml. Fluorescence microscopy and SEM demonstrated the nanoparticles' capacity to prevent bacterial surface adhesion. EDX research revealed that the organic extract efficiently formed silver nanoparticles with considerable oxygen incorporation, which was attributed to phytochemicals that stabilize AgNPs and prevent accumulation. FTIR spectroscopy indicated the existence of hydroxyl, carbonyl, and carboxylate groups, which are essential for nanoparticle stability. TEM revealed that the AgNPs were spheroidal, with diameters ranging from 40 to 60 nm and an average of 46 nm. These results demonstrate the efficacy of H. speciosa extract in creating stable, well-defined AgNPs suited for a variety of applications. This work underlines the potential of green-synthesized AgNPs in biomedical applications, notably in the treatment of S. aureus biofilm-associated illnesses. The thorough characterization gives important information on the stability and efficiency of these biogenic nanoparticles.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biotechnology, St. Mary's College (Autonomous), Thrissur, Kerala, 680020, India; Manipur International University, Imphal, Manipur, 795140, India.
| | - Janarthanam Hemapriya
- Department of Microbiology, DKM College for Women, Vellore, Tamil Nadu, 632001, India
| | | | - Kuppusamy Sathishkumar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, Tamil Nadu, 602105, India.
| | | |
Collapse
|
3
|
Murugan U, Gusain D, Balasubramani B, Srivastava S, Ganesh S, Ambattu Raghavannambiar S, Ramaraj K. A comprehensive review of environment-friendly biomimetic bionic superhydrophobic surfaces. BIOFOULING 2024:1-23. [PMID: 39422280 DOI: 10.1080/08927014.2024.2414922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
Marine fouling is a global problem that harms the ocean's ecosystem and the marine industrial sector. Traditional antifouling methods use harmful agents that damage the environment. As a result, recent research has focused on developing environmentally friendly, long-lasting, and sustainable antifouling solutions. Scientists have turned to nature for inspiration, particularly the water-repellent properties found in the microstructures of plants, insects and animals like the lotus leaf, butterfly, and shark. This review summarizes the current trends in developing superhydrophobic materials and fabrication techniques for bionic antifouling strategies. These strategies mimic the surface microstructures of various biological species, including the lotus leaf, coral tentacles, and the skins of sharks, whales, and dolphins. The review also discusses the technological applications of these biomimetic materials and the challenges associated with implementing them in the marine sector. Overall, the goal is to harness the superhydrophobicity of natural surfaces to create effective antifouling solutions.
Collapse
Affiliation(s)
- Udhayakumar Murugan
- Department of Aerospace Engineering, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore, India
| | - Dakshesh Gusain
- Department of Aerospace Engineering, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore, India
| | - Baskar Balasubramani
- Department of Aerospace Engineering, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore, India
| | - Sagar Srivastava
- Department of Aerospace Engineering, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore, India
| | - Sai Ganesh
- Department of Aerospace Engineering, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore, India
| | | | - Kannan Ramaraj
- Department of Aerospace Engineering, Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore, India
| |
Collapse
|
4
|
D’Angelo C, Faggiano S, Imbimbo P, Viale E, Casillo A, Bettati S, Olimpo D, Tutino ML, Monti DM, Corsaro MM, Ronda L, Parrilli E. Pentadecanoic Acid-Releasing PDMS: Towards a New Material to Prevent S. epidermidis Biofilm Formation. Int J Mol Sci 2024; 25:10727. [PMID: 39409056 PMCID: PMC11476977 DOI: 10.3390/ijms251910727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Microbial biofilm formation on medical devices paves the way for device-associated infections. Staphylococcus epidermidis is one of the most common strains involved in such infections as it is able to colonize numerous devices, such as intravenous catheters, prosthetic joints, and heart valves. We previously reported the antibiofilm activity against S. epidermidis of pentadecanoic acid (PDA) deposited by drop-casting on the silicon-based polymer poly(dimethyl)siloxane (PDMS). This material exerted an antibiofilm activity by releasing PDA; however, a toxic effect on bacterial cells was observed, which could potentially favor the emergence of resistant strains. To develop a PDA-functionalized material for medical use and overcome the problem of toxicity, we produced PDA-doped PDMS by either spray-coating or PDA incorporation during PDMS polymerization. Furthermore, we created a strategy to assess the kinetics of PDA release using ADIFAB, a very sensitive free fatty acids fluorescent probe. Spray-coating resulted in the most promising strategy as the concentration of released PDA was in the range 0.8-1.5 μM over 21 days, ensuring long-term effectiveness of the antibiofilm molecule. Moreover, the new coated material resulted biocompatible when tested on immortalized human keratinocytes. Our results indicate that PDA spray-coated PDMS is a promising material for the production of medical devices endowed with antibiofilm activity.
Collapse
Affiliation(s)
- Caterina D’Angelo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Serena Faggiano
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124 Parma, Italy
- Institute of Biophysics, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (S.B.); (L.R.)
| | - Paola Imbimbo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Elisabetta Viale
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Angela Casillo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Stefano Bettati
- Institute of Biophysics, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (S.B.); (L.R.)
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Diana Olimpo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Daria Maria Monti
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Maria Michela Corsaro
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Luca Ronda
- Institute of Biophysics, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (S.B.); (L.R.)
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| |
Collapse
|
5
|
Salazar-Sesatty HA, Montoya-Hinojosa EI, Villarreal-Salazar V, Alvizo-Baez CA, Camacho-Ortiz A, Terrazas-Armendariz LD, Luna-Cruz IE, Alcocer-González JM, Villarreal-Treviño L, Flores-Treviño S. Biofilm Eradication and Inhibition of Methicillin-Resistant Staphylococcus Clinical Isolates by Curcumin-Chitosan Magnetic Nanoparticles. Jpn J Infect Dis 2024; 77:260-268. [PMID: 38825455 DOI: 10.7883/yoken.jjid.2024.034] [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: 06/04/2024]
Abstract
Biofilm-producing methicillin-resistant Staphylococcus aureus (MRSA) and coagulase-negative staphylococci (MR-CoNS) pose clinical challenges in treating healthcare-associated infections. As alternative antimicrobial options are needed, in this study, we aimed to determine the effect of curcumin-chitosan magnetic nanoparticles (Cur-Chi-MNP) on the biofilms of staphylococcal clinical isolates. MRSA and CoNS clinical isolates were identified using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Antimicrobial susceptibility testing was performed using the broth microdilutions. Nanoparticles were synthesized by the co-precipitation of magnetic nanoparticles (MNP) and encapsulated by the ionotropic gelation of curcumin (Cur) and chitosan (Chi). Biofilm inhibition and eradication by nanoparticles, with and without the addition of oxacillin (OXA), were assessed in Staphylococcus strains. Cur-Chi-MNP showed antimicrobial activity against planktonic cells of MRSA and MR-CoNS strains and inhibited MRSA biofilm. The addition of OXA to Cur-Chi-MNP increased the biofilm inhibition and eradication activity against all staphylococcal strains (P = 0.0007), and higher biofilm activity was observed in the early biofilm stages. Cur-Chi-MNP showed antimicrobial and biofilm inhibitory activities against S. aureus. Addition of OXA increased biofilm inhibition and eradication activity against all staphylococcal strains. A combination treatment of Cur-Chi-MNP and OXA could potentially be used to treat staphylococcal biofilm-associated infections in the early stages before the establishment of biofilm bacterial cells.
Collapse
Affiliation(s)
| | | | | | | | - Adrián Camacho-Ortiz
- Department of Infectious Diseases, University Hospital Dr. José Eleuterio González and School of Medicine, Autonomous University of Nuevo Leon, Mexico
| | | | | | | | - Licet Villarreal-Treviño
- Departament of Microbiology, School of Biological Sciences, Autonomous University of Nuevo Leon, Mexico
| | - Samantha Flores-Treviño
- Department of Infectious Diseases, University Hospital Dr. José Eleuterio González and School of Medicine, Autonomous University of Nuevo Leon, Mexico
| |
Collapse
|
6
|
Ma X, Wu Z, Li J, Yang Y. Functional Study of desKR: a Lineage-Specific Two-Component System Positively Regulating Staphylococcus aureus Biofilm Formation. Infect Drug Resist 2024; 17:4037-4053. [PMID: 39309069 PMCID: PMC11416117 DOI: 10.2147/idr.s485049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Purpose Biofilms significantly contribute to the persistence and antibiotic resistance of Staphylococcus aureus infections. However, the regulatory mechanisms governing biofilm formation of S. aureus remain not fully elucidated. This study aimed to investigate the function of the S. aureus lineage-specific two-component system, desKR, in biofilm regulation and pathogenicity. Methods Bioinformatic analysis was conducted to assess the prevalence of desKR across various S. aureus lineages and to examine its structural features. The impact of desKR on S. aureus pathogenicity was evaluated using in vivo mouse models, including skin abscess, bloodstream infection, and nasal colonization models. Crystal violet staining and confocal laser scanning microscopy were utilized to examine the impact of desKR on S. aureus biofilm formation. Mechanistic insights into desKR-mediated biofilm regulation were investigated by quantifying polysaccharide intercellular adhesin (PIA) production, extracellular DNA (eDNA) release, autolysis assays, and RT-qPCR. Results The prevalence of desKR varied among different S. aureus lineages, with notably low carriage rates in ST398 and ST59 lineages. Deletion of desKR in NCTC8325 strain resulted in decreased susceptibility to β-lactam and glycopeptide antibiotics. Although desKR did not significantly affect acute pathogenicity, the ΔdesKR mutant exhibited significantly reduced nasal colonization and biofilm-forming ability. Overexpression of desKR in naturally desKR-lacking strains (ST398 and ST59) enhanced biofilm formation, suggesting a lineage-independent effect. Phenotypic assays further revealed that the ΔdesKR mutant showed reduced PIA production, decreased eDNA release, and lower autolysis rates. RT-qPCR indicated significant downregulation of icaA, icaD, icaB, and icaC genes, along with upregulation of icaR, whereas autolysis-related genes remained unchanged. Conclusion The desKR two-component system positively regulates S. aureus biofilm formation in a lineage-independent manner, primarily by modulating PIA synthesis via the ica operon. These findings provide new insights into the molecular mechanisms of biofilm formation in S. aureus and highlight desKR as a potential target for therapeutic strategies aimed at combating biofilm-associated infections.
Collapse
Affiliation(s)
- Xinyan Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| | - Ziyan Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| | - Junpeng Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| | - Yang Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| |
Collapse
|
7
|
Jandl B, Dighe S, Gasche C, Makristathis A, Muttenthaler M. Intestinal biofilms: pathophysiological relevance, host defense, and therapeutic opportunities. Clin Microbiol Rev 2024; 37:e0013323. [PMID: 38995034 PMCID: PMC11391705 DOI: 10.1128/cmr.00133-23] [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] [Indexed: 07/13/2024] Open
Abstract
SUMMARYThe human intestinal tract harbors a profound variety of microorganisms that live in symbiosis with the host and each other. It is a complex and highly dynamic environment whose homeostasis directly relates to human health. Dysbiosis of the gut microbiota and polymicrobial biofilms have been associated with gastrointestinal diseases, including irritable bowel syndrome, inflammatory bowel diseases, and colorectal cancers. This review covers the molecular composition and organization of intestinal biofilms, mechanistic aspects of biofilm signaling networks for bacterial communication and behavior, and synergistic effects in polymicrobial biofilms. It further describes the clinical relevance and diseases associated with gut biofilms, the role of biofilms in antimicrobial resistance, and the intestinal host defense system and therapeutic strategies counteracting biofilms. Taken together, this review summarizes the latest knowledge and research on intestinal biofilms and their role in gut disorders and provides directions toward the development of biofilm-specific treatments.
Collapse
Affiliation(s)
- Bernhard Jandl
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Satish Dighe
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christoph Gasche
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- Loha for Life, Center for Gastroenterology and Iron Deficiency, Vienna, Austria
| | - Athanasios Makristathis
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
8
|
Çali A, Çelik C. Determination of in vitro synergy and antibiofilm activities of antimicrobials and essential oil components. BIOFOULING 2024; 40:483-498. [PMID: 39069795 DOI: 10.1080/08927014.2024.2381587] [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: 03/29/2024] [Revised: 06/14/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
Using existing adrentimicrobials with essential oil components to prevent antimicrobial resistance is an alternative strategy. This study aimed to evaluate the resistance status, synergistic combinations, and in vitro biofilm formation activities of clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA), Stenotrophomonas maltophilia and Candida albicans against antimicrobial agents and cinnamaldehyde, carvacrol, eugenol, limonene and eucalyptol. Antimicrobial activities were evaluated by microdilution, cytotoxicity by XTT, synergy by checkerboard and time-kill, and biofilm inhibition by microplate methods. Cinnamaldehyde and carvacrol showed strong antimicrobial activity. Synergistic effects were observed when using all essential oils with antimicrobials. Only two C. albicans isolates showed antagonism with cinnamaldehyde and fluconazole. The constituents showed cytotoxic effects in the L929 cell line (except limonene). A time-kill analysis revealed a bacteriostatic effect on S. maltophilia and MRSA isolates and a fungicidal effect on C. albicans isolates. These results are important for further research to improve antimicrobial efficacy or to develop new agents.
Collapse
Affiliation(s)
- Abdulhamit Çali
- Medical Laboratory Techniques, Vocational School of Health Services, Lokman Hekim University, Ankara, Turkey
| | - Cem Çelik
- Department of Medical Microbiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
9
|
Leistikow KR, May DS, Suh WS, Vargas Asensio G, Schaenzer AJ, Currie CR, Hristova KR. Bacillus subtilis-derived peptides disrupt quorum sensing and biofilm assembly in multidrug-resistant Staphylococcus aureus. mSystems 2024; 9:e0071224. [PMID: 38990088 PMCID: PMC11334493 DOI: 10.1128/msystems.00712-24] [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: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 07/12/2024] Open
Abstract
Multidrug-resistant Staphylococcus aureus is one of the most clinically important pathogens in the world, with infections leading to high rates of morbidity and mortality in both humans and animals. The ability of S. aureus to form biofilms protects cells from antibiotics and promotes the transfer of antibiotic resistance genes; therefore, new strategies aimed at inhibiting biofilm growth are urgently needed. Probiotic species, including Bacillus subtilis, are gaining interest as potential therapies against S. aureus for their ability to reduce S. aureus colonization and virulence. Here, we search for strains and microbially derived compounds with strong antibiofilm activity against multidrug-resistant S. aureus by isolating and screening Bacillus strains from a variety of agricultural environments. From a total of 1,123 environmental isolates, we identify a single strain B. subtilis 6D1, with a potent ability to inhibit biofilm growth, disassemble mature biofilm, and improve antibiotic sensitivity of S. aureus biofilms through an Agr quorum sensing interference mechanism. Biochemical and molecular networking analysis of an active organic fraction revealed multiple surfactin isoforms, and an uncharacterized peptide was driving this antibiofilm activity. Compared with commercial high-performance liquid chromatography grade surfactin obtained from B. subtilis, we show these B. subtilis 6D1 peptides are significantly better at inhibiting biofilm formation in all four S. aureus Agr backgrounds and preventing S. aureus-induced cytotoxicity when applied to HT29 human intestinal cells. Our study illustrates the potential of exploring microbial strain diversity to discover novel antibiofilm agents that may help combat multidrug-resistant S. aureus infections and enhance antibiotic efficacy in clinical and veterinary settings. IMPORTANCE The formation of biofilms by multidrug-resistant bacterial pathogens, such as Staphylococcus aureus, increases these microorganisms' virulence and decreases the efficacy of common antibiotic regimens. Probiotics possess a variety of strain-specific strategies to reduce biofilm formation in competing organisms; however, the mechanisms and compounds responsible for these phenomena often go uncharacterized. In this study, we identified a mixture of small probiotic-derived peptides capable of Agr quorum sensing interference as one of the mechanisms driving antibiofilm activity against S. aureus. This collection of peptides also improved antibiotic killing and protected human gut epithelial cells from S. aureus-induced toxicity by stimulating an adaptive cytokine response. We conclude that purposeful strain screening and selection efforts can be used to identify unique probiotic strains that possess specially desired mechanisms of action. This information can be used to further improve our understanding of the ways in which probiotic and probiotic-derived compounds can be applied to prevent bacterial infections or improve bacterial sensitivity to antibiotics in clinical and agricultural settings.
Collapse
Affiliation(s)
- Kyle R. Leistikow
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Daniel S. May
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Chemistry, Washington College, Chestertown, Maryland, USA
| | - Won Se Suh
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Adam J. Schaenzer
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Cameron R. Currie
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
10
|
Purgato GA, Píccolo MS, Moreira MAS, Pizziolo VR, Diaz-Muñoz G, Rossi CC, Diaz MAN. Isolation and identification of antimicrobial multicyclic terpenoids from the medicinal plant Salvia officinalis and development of a formulation against clinical Staphylococcus aureus strains. Lett Appl Microbiol 2024; 77:ovae077. [PMID: 39127610 DOI: 10.1093/lambio/ovae077] [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: 05/20/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Staphylococcus aureus, particularly multi-drug resistant strains, presents significant challenges in dairy farming due to its role in causing bovine mastitis, which leads to substantial economic losses and limited treatment options. Seeking alternative therapies, we investigated the potential of a topical formulation derived from the medicinal herb Salvia officinalis to combat S. aureus growth and biofilms associated with bovine mastitis. Through systematic extraction in different solvents and fractionation by column chromatography, we isolated and identified three key multicyclic terpenoids-ferruginol, sugiol, and sclareol-exhibiting significant antimicrobial activity. The formulation effectively inhibited biofilm formation, with minimum inhibitory concentration (MIC) values ranging from 0.09 to 0.74 mg ml-1 against clinical S. aureus strains, comparable to or lower than those of the pure compounds. Moreover, it displayed robust anti-adhesive properties, reducing biofilm formation by 20%-79% at subinhibitory concentrations. Furthermore, the formulation successfully disrupted pre-existing biofilms, achieving reductions ranging from 30% to 82%. Cytotoxicity assays confirmed the safety of the formulation on mammary epithelial cells, with cell viability maintained at 100% at MIC. Our findings underscore the therapeutic potential of Sa. officinalis-derived compounds in managing bovine mastitis caused by S. aureus, emphasizing their antimicrobial efficacy and safety profile.
Collapse
Affiliation(s)
| | - Mayra Soares Píccolo
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, MG 36570-900, Brazil
| | | | - Virgínia Ramos Pizziolo
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, MG 36570-900, Brazil
| | - Gaspar Diaz-Muñoz
- Departamento de Química, Universidade Federal de Minas Gerais, MG 31270-901, Brazil
| | - Ciro César Rossi
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, MG 36570-900, Brazil
| | | |
Collapse
|
11
|
Power AD, Mok WWK. Agar and agarose used for Staphylococcus aureus biofilm cultivation impact fluoroquinolone tolerance. J Appl Microbiol 2024; 135:lxae191. [PMID: 39066496 DOI: 10.1093/jambio/lxae191] [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: 05/29/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024]
Abstract
AIMS Staphylococcus aureus is an opportunistic pathogen whose treatment is further complicated by its ability to form biofilms. In this study, we examine the impact of growing S. aureus biofilms on different polymerizing surfaces, specifically agar and agarose, on the pathogen's tolerance to fluoroquinolones. METHODS AND RESULTS Biofilms of two methicillin-resistant strains of S. aureus were grown on agar or agarose in the presence of the same added nutrients, and their antibiotic susceptibility to two fluoroquinolones, moxifloxacin (MXF) and delafloxacin (DLX), were measured. We also compared the metabolism and extracellular polymeric substances (EPS) production of biofilms that were grown on agar and agarose. CONCLUSIONS Biofilms that were grown on agarose were consistently more susceptible to antibiotics than those grown on agar. We found that in biofilms that were grown on agar, extracellular protein composition was higher, and adding EPS to agarose-grown biofilms increased their tolerance to DLX to levels that were comparable to agar-grown biofilms.
Collapse
Affiliation(s)
- Angela D Power
- Department of Molecular Biology & Biophysics, UConn Health, Farmington, CT 06032, United States
| | - Wendy W K Mok
- Department of Molecular Biology & Biophysics, UConn Health, Farmington, CT 06032, United States
| |
Collapse
|
12
|
Khodabux RMJ, Mariappan S, Sekar U. Virulence, Susceptibility Profile, and Clinical Characteristics of Pathogenic Coagulase-Negative Staphylococci. Cureus 2024; 16:e66397. [PMID: 39247021 PMCID: PMC11379411 DOI: 10.7759/cureus.66397] [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] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Background Coagulase-negative staphylococci (CoNS) are emerging as clinically significant pathogens. A high proportion of methicillin resistance along with intense biofilm-producing ability render CoNS-related infections challenging to treat. This study was undertaken to investigate the mechanisms of methicillin resistance, identify genes encoding for virulence, and their association with clinical outcomes among clinical isolates of Staphylococci in a tertiary care center. Methods A total of 203 clinical isolates were included in this study. Susceptibility to various antibiotics was determined by the disc diffusion method. Methicillin resistance was screened using cefoxitin disc, mecA and mecC genes were detected using polymerase chain reaction (PCR). PCR was performed to detect five virulence genes: atlE, aap, fbe, embp, and icaAB. Staphylococcal cassette chromosome mec (SCCmec) types were identified by multiplex PCR. Statistical analysis was performed using SPSS software (IBM Inc., Armonk, New York). The Chi-squared test was used to compare the distribution of virulence genes among methicillin-susceptible resistant CoNS. A p-value of less than 0.5 was considered significant. Results In the current study, 60% (122/203) of CoNS were methicillin-resistant, and SCCmec type I was the most common. Among the 203 CoNS, 24.6% (50/203) isolates harbored one or more virulence genes in them. Conclusion CoNS have relatively low virulence as only 24.6% of isolates carried the virulence genes. Nevertheless, the variety of diseases linked to these species indicates the necessity for accurate identification and precise reporting of antimicrobial susceptibility to avoid adverse outcomes.
Collapse
Affiliation(s)
| | - Shanthi Mariappan
- Microbiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Uma Sekar
- Microbiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| |
Collapse
|
13
|
Namuga C, Muwonge H, Nasifu K, Sekandi P, Sekulima T, Kirabira JB. Hoslundia opposita vahl; a potential source of bioactive compounds with antioxidant and antibiofilm activity for wound healing. BMC Complement Med Ther 2024; 24:236. [PMID: 38886717 PMCID: PMC11181642 DOI: 10.1186/s12906-024-04540-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Biofilms and oxidative stress retard wound healing. The resistance of biofilms to antibiotics has led to a search for alternative approaches in biofilm elimination. Antioxidants work synergistically with antibacterial agents against biofilms. Hence recent research has suggested plants as candidates in the development of new alternatives in biofilm treatments and as antioxidants due to the presence of phytocompounds which are responsible for their bioactivities. Hoslundia opposita Vahl is one of the plants used by traditional healers to treat wounds and other infections, this makes it a potential candidate for drug discovery hence, in this study, we investigate the antibiofilm and antioxidant activity of methanolic extract of hoslundia opposita Vahl from Uganda. We also identify phytochemicals responsible for its bioactivity. METHOD the plant was extracted by maceration using methanol, and the extract was investigated for antioxidant activity using 2,2-diphenyl-1-picrylhydrazyl radical (DPPH) assay. The antibiofilm activity using microtiter plate assay (MTP) assay where the Minimum biofilm inhibitory concentration required to inhibit 50% or 90% of the biofilm (MBIC50 and MBIC90) and Minimum biofilm eradication concentration required to remove 50% or 90% of the biofilm (MBEC50 and MBEC90) were measured. It was further analysed for its phytochemical composition using quantitative screening, as well as Gas chromatography-mass spectrometry (GC-MS) and Liquid chromatography mass-spectrometry (LC-MS). RESULTS H. Opposita Vahl extract showed good antioxidant activity with of 249.6 mg/mL. It inhibited the growth of P. aeruginosa and S. aureus biofilms with MBIC50 of 28.37 mg/mL and 10 mg/mL, respectively. It showed the ability to eradicate P. aeruginosa and S. aureus biofilms with MBEC50 of 23.85 and 39.01 mg/mL respectively. Phytochemical analysis revealed the presence of alkaloids, tannins, flavonoids, and phenols. GC-MS analysis revealed 122 compounds in the extract of which, 23 have evidence of antioxidant or antibiofilm activity in literature. The most abundant compounds were; 1,4- Citric acid, Tetracontane-1,40-diol (43.43.3%, 1, Olean-12-en-28-oic acid, 3-hydroxy-, methyl ester, (3.beta) (15.36%) 9-Octadecenamide (12.50%), Squalene (11.85%) Palmitic Acid 4TMS (11.28%), and alpha Amyrin (11.27%). The LC-MS identified 115 and 57 compounds in multiple reaction mode (MRM) and scan modes respectively. CONCLUSION H. opposita Vahl showed antibiofilm and antioxidant activity due to bioactive compounds identified, hence the study justifies its use for wound healing. It can be utilised in further development of new drugs as antibiofilm and antioxidants.
Collapse
Affiliation(s)
- Catherine Namuga
- Depatment of Polymer, Textile, and Industrial Engineering, Busitema University, P. O. Box 256, Tororo, Uganda.
- Department of Physiology, College of Health Sciences, Makerere University, P. O. Box 7062, Kampala, Uganda.
| | - Haruna Muwonge
- Department of Chemistry, College of Natural Sciences, Makerere University, P. O. Box 7062, Kampala, Uganda
| | - Kerebba Nasifu
- Department of Microbiology, College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P. O. Box 7062, Kampala, Uganda
| | - Peter Sekandi
- Department of Microbiology, College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P. O. Box 7062, Kampala, Uganda
| | - Tahalu Sekulima
- Department of Mechanical Engineering, College of Engineering, Design, Art, and Technology, Makerere University, Kampala, Uganda
| | - John Baptist Kirabira
- Department of Physiology, College of Health Sciences, Makerere University, P. O. Box 7062, Kampala, Uganda
| |
Collapse
|
14
|
Yu G, Xi H, Sheng T, Lin J, Luo Z, Xu J. Sub-inhibitory concentrations of tetrabromobisphenol A induce the biofilm formation of methicillin-resistant Staphylococcus aureus. Arch Microbiol 2024; 206:301. [PMID: 38874781 DOI: 10.1007/s00203-024-04022-3] [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: 04/19/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
Biofilm formation by methicillin-resistant Staphylococcus aureus (MRSA) on indwelling medical devices complicates the treatment of infection. Tetrabromobisphenol A (TBBPA), a synthetic, lipophilic, halogenated aromatic compound widely used as an additive in plastics and electronic products, has raised environmental concerns due to its potential for bioaccumulation. This study investigated the impact of sub-inhibitory concentrations of TBBPA on MRSA biofilm formation. Crystal violet staining and confocal laser scanning microscopy analysis demonstrated that 1/8 MIC (0.5 µg/mL) of TBBPA significantly stimulated MRSA biofilm formation (P < 0.0001). MTT assays indicated that the metabolic activity within the biofilms increased by 15.60-40.85% compared to untreated controls. Dot blot immunoassay, autolysis assay, and extracellular DNA (eDNA) quantification further revealed TBBPA enhanced the production of polysaccharide intercellular adhesin (PIA) and eDNA, which are key biofilm components. Additionally, TBBPA was found to enhance the production of staphyloxanthin, facilitating MRSA survival under oxidative conditions and in human whole blood. RT-qPCR analysis showed that TBBPA significantly upregulated genes associated with biofilm formation (icaA, atlA, sarA), staphyloxanthin biosynthesis (crtM and sigB), and oxidative stress responses (sodA and katA). These findings suggest that TBBPA promotes MRSA biofilm development and enhances bacterial resistance to adverse conditions, thereby potentially exacerbating risks to human health.
Collapse
Affiliation(s)
- Guofang Yu
- Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Huimin Xi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Tianle Sheng
- Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jin Lin
- Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Zhaoxia Luo
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| | - Jianqing Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
15
|
Acosta RB, Durantini EN, Spesia MB. Evaluation of quantification methods to determine photodynamic action on mono- and dual-species bacterial biofilms. Photochem Photobiol Sci 2024; 23:1195-1208. [PMID: 38703274 DOI: 10.1007/s43630-024-00586-7] [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: 01/22/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
The effect of photodynamic inactivation (PDI) sensitized by 5,10,15,20-tetra(4-N,N,N-trimethylammoniophenyl)porphyrin (TMAP4+) on different components of mono- and dual-species biofilms of Staphylococcus aureus and Escherichia coli was determined by different methods. First, the plate count technique showed that TMAP4+-PDI was more effective on S. aureus than E. coli biofilm. However, crystal violet staining revealed no significant differences between before and after PDI biofilms of both bacteria. On the other hand, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method indicated a reduction in viable cells as the light exposure time increases in both, mono- and dual-species biofilms. Furthermore, it was determined that as the irradiation time increases, the amount of extracellular polymeric substances present in the biofilms decreased. This effect was presented in both strains and in the mixed biofilm, being more evident in S. aureus mono-specie biofilm. Finally, scanning electron microscopy analysis showed a decrease in the number of cells forming the biofilm after photosensitization treatments. This information makes it possible to determine whether the photodynamic action is based on damage to metabolic activity, extracellular matrix and/or biomass, which may be useful in establishing a fully effective PDI protocol for the treatment of microorganisms growing as biofilms.
Collapse
Affiliation(s)
- Rocío B Acosta
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, X5804BYA, Río Cuarto, Córdoba, Argentina
| | - Edgardo N Durantini
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, X5804BYA, Río Cuarto, Córdoba, Argentina
| | - Mariana B Spesia
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, X5804BYA, Río Cuarto, Córdoba, Argentina.
| |
Collapse
|
16
|
Garrine M, Andrade M, Neves J, Mandomando I, Couto I, Costa SS. Exploring the virulence potential of Staphylococcus aureus CC121 and CC152 lineages related to paediatric community-acquired bacteraemia in Manhiça, Mozambique. Sci Rep 2024; 14:10758. [PMID: 38730020 PMCID: PMC11087594 DOI: 10.1038/s41598-024-61345-3] [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: 12/19/2023] [Accepted: 05/05/2024] [Indexed: 05/12/2024] Open
Abstract
Staphylococcus aureus is a frequent agent of bacteraemia. This bacterium has a variety of virulence traits that allow the establishment and maintenance of infection. This study explored the virulence profile of S. aureus strains causing paediatric bacteraemia (SAB) in Manhiça district, Mozambique. We analysed 336 S. aureus strains isolated from blood cultures of children younger than 5 years admitted to the Manhiça District Hospital between 2001 and 2019, previously characterized for antibiotic susceptibility and clonality. The strains virulence potential was evaluated by PCR detection of the Panton-Valentine leucocidin (PVL) encoding genes, lukS-PV/lukF-PV, assessment of the capacity for biofilm formation and pathogenicity assays in Galleria mellonella. The overall carriage of PVL-encoding genes was over 40%, although reaching ~ 70 to 100% in the last years (2014 to 2019), potentially linked to the emergence of CC152 lineage. Strong biofilm production was a frequent trait of CC152 strains. Representative CC152 and CC121 strains showed higher virulence potential in the G. mellonella model when compared to reference strains, with variations within and between CCs. Our results highlight the importance of monitoring the emergent CC152-MSSA-PVL+ and other lineages, as they display important virulence traits that may negatively impact the management of SAB paediatric patients in Manhiça district, Mozambique.
Collapse
Affiliation(s)
- Marcelino Garrine
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Mariana Andrade
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Joana Neves
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Inácio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
- ISGlobal-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Isabel Couto
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Sofia Santos Costa
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| |
Collapse
|
17
|
Lopes N, Pereira RB, Correia A, Vilanova M, Cerca N, França A. Deletion of codY impairs Staphylococcus epidermidis biofilm formation, generation of viable but non-culturable cells and stimulates cytokine production in human macrophages. J Med Microbiol 2024; 73. [PMID: 38743043 DOI: 10.1099/jmm.0.001837] [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: 05/16/2024] Open
Abstract
Introduction. Staphylococcus epidermidis biofilms are one of the major causes of bloodstream infections related to the use of medical devices. The diagnosis of these infections is challenging, delaying their treatment and resulting in increased morbidity and mortality rates. As such, it is urgent to characterize the mechanisms employed by this bacterium to endure antibiotic treatments and the response of the host immune system, to develop more effective therapeutic strategies. In several bacterial species, the gene codY was shown to encode a protein that regulates the expression of genes involved in biofilm formation and immune evasion. Additionally, in a previous study, our group generated evidence indicating that codY is involved in the emergence of viable but non-culturable (VBNC) cells in S. epidermidis.Gap statement/Hypothesis. As such, we hypothesized that the gene codY has have an important role in this bacterium virulence.Aim. This study aimed to assess, for the first time, the impact of the deletion of the gene codY in S. epidermidis virulence, namely, in antibiotic susceptibility, biofilm formation, VBNC state emergence and in vitro host immune system response.Methodology. Using an allelic replacement strategy, we constructed and then characterized an S. epidermidis strain lacking codY, in regards to biofilm and VBNC cell formation, susceptibility to antibiotics as well as their role in the interaction with human blood and plasma. Additionally, we investigate whether the codY gene can impact the activation of innate immune cells by evaluating the production of both pro- and anti-inflammatory cytokines by THP-1 macrophages.Results. We demonstrated that the deletion of the gene codY resulted in biofilms with less c.f.u. counts and fewer VBNC cells. Furthermore, we show that although WT and mutant cells were similarly internalized in vitro by human macrophages, a stronger cytokine response was elicited by the mutant in a toll-like receptor 4-dependent manner.Conclusion. Our results indicate that codY contributes to S. epidermidis virulence, which in turn may have an impact on our ability to manage the biofilm-associated infections caused by this bacterium.
Collapse
Affiliation(s)
- Nathalie Lopes
- Laboratório de Investigação em Biofilmes Rosário Oliveira (LIBRO), Centro de Engenharia Biológica (CEB), Universidade do Minho, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Renato B Pereira
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Alexandra Correia
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Manuel Vilanova
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Nuno Cerca
- Laboratório de Investigação em Biofilmes Rosário Oliveira (LIBRO), Centro de Engenharia Biológica (CEB), Universidade do Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- LABBELS-Laboratório Associado, Braga, Guimarães, Portugal
| | - Angela França
- Laboratório de Investigação em Biofilmes Rosário Oliveira (LIBRO), Centro de Engenharia Biológica (CEB), Universidade do Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- LABBELS-Laboratório Associado, Braga, Guimarães, Portugal
| |
Collapse
|
18
|
Aboelnaga N, Elsayed SW, Abdelsalam NA, Salem S, Saif NA, Elsayed M, Ayman S, Nasr M, Elhadidy M. Deciphering the dynamics of methicillin-resistant Staphylococcus aureus biofilm formation: from molecular signaling to nanotherapeutic advances. Cell Commun Signal 2024; 22:188. [PMID: 38519959 PMCID: PMC10958940 DOI: 10.1186/s12964-024-01511-2] [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: 11/20/2023] [Accepted: 02/01/2024] [Indexed: 03/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a global threat, necessitating the development of effective solutions to combat this emerging superbug. In response to selective pressures within healthcare, community, and livestock settings, MRSA has evolved increased biofilm formation as a multifaceted virulence and defensive mechanism, enabling the bacterium to thrive in harsh conditions. This review discusses the molecular mechanisms contributing to biofilm formation across its developmental stages, hence representing a step forward in developing promising strategies for impeding or eradicating biofilms. During staphylococcal biofilm development, cell wall-anchored proteins attach bacterial cells to biotic or abiotic surfaces; extracellular polymeric substances build scaffolds for biofilm formation; the cidABC operon controls cell lysis within the biofilm, and proteases facilitate dispersal. Beside the three main sequential stages of biofilm formation (attachment, maturation, and dispersal), this review unveils two unique developmental stages in the biofilm formation process for MRSA; multiplication and exodus. We also highlighted the quorum sensing as a cell-to-cell communication process, allowing distant bacterial cells to adapt to the conditions surrounding the bacterial biofilm. In S. aureus, the quorum sensing process is mediated by autoinducing peptides (AIPs) as signaling molecules, with the accessory gene regulator system playing a pivotal role in orchestrating the production of AIPs and various virulence factors. Several quorum inhibitors showed promising anti-virulence and antibiofilm effects that vary in type and function according to the targeted molecule. Disrupting the biofilm architecture and eradicating sessile bacterial cells are crucial steps to prevent colonization on other surfaces or organs. In this context, nanoparticles emerge as efficient carriers for delivering antimicrobial and antibiofilm agents throughout the biofilm architecture. Although metal-based nanoparticles have been previously used in combatting biofilms, its non-degradability and toxicity within the human body presents a real challenge. Therefore, organic nanoparticles in conjunction with quorum inhibitors have been proposed as a promising strategy against biofilms. As nanotherapeutics continue to gain recognition as an antibiofilm strategy, the development of more antibiofilm nanotherapeutics could offer a promising solution to combat biofilm-mediated resistance.
Collapse
Affiliation(s)
- Nirmeen Aboelnaga
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Salma W Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nehal Adel Abdelsalam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Salma Salem
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Nehal A Saif
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Manar Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Shehab Ayman
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Elhadidy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt.
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
19
|
Dsouza FP, Dinesh S, Sharma S. Understanding the intricacies of microbial biofilm formation and its endurance in chronic infections: a key to advancing biofilm-targeted therapeutic strategies. Arch Microbiol 2024; 206:85. [PMID: 38300317 DOI: 10.1007/s00203-023-03802-7] [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: 11/10/2023] [Revised: 12/04/2023] [Accepted: 12/16/2023] [Indexed: 02/02/2024]
Abstract
Bacterial biofilms can adhere to various surfaces in the environment with human beings being no exception. Enclosed in a self-secreted matrix which contains extracellular polymeric substances, biofilms are intricate communities of bacteria that play a significant role across various sectors and raise concerns for public health, medicine and industries. These complex structures allow free-floating planktonic cells to adopt multicellular mode of growth which leads to persistent infections. This is of great concern as biofilms can withstand external attacks which include antibiotics and immune responses. A more comprehensive and innovative approach to therapy is needed in view of the increasing issue of bacterial resistance brought on by the overuse of conventional antimicrobial medications. Thus, to oppose the challenges posed by biofilm-related infections, innovative therapeutic strategies are being explored which include targeting extracellular polymeric substances, quorum sensing, and persister cells. Biofilm-responsive nanoparticles show promising results by improving drug delivery and reducing the side effects. This review comprehensively examines the factors influencing biofilm formation, host immune defence mechanisms, infections caused by biofilms, diagnostic approaches, and biofilm-targeted therapies.
Collapse
Affiliation(s)
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India.
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru, Karnataka, 560043, India
| |
Collapse
|
20
|
Bilgin M, Dosler S, Otuk G. Antibiotic adjuvant activities of quorum sensing signal molecules DSF and BDSF against mature biofilms of Staphylococci. J Chemother 2024; 36:11-23. [PMID: 37873740 DOI: 10.1080/1120009x.2023.2270743] [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: 01/21/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
Among promising antibiofilm compounds, quorum-sensing (QS) molecules that regulate biological processes such as biofilm formation and intra- or interspecies communication appear to be good candidates. The invitro antibiotic-adjuvant effects of QS molecules diffusible signal factor (DSF) and B. cenocepacia producing-DSF (BDSF) were investigated against mature Staphylococcal biofilms. Broth microdilution methods were used for the determinations of MIC, MBC, MBIC, and MBEC, and bactericidal activities were determined by TKC method. The lowest MICs were obtained with ciprofloxacin and gentamicin, and MBECs with ciprofloxacin. DSF and BDSF at 0.5 µM decreased the MICs as 2-8, and 2-32 fold, respectively. In TKC studies, -cidal activities were achieved by BDSF + gentamycin, or ciprofloxacin, and DSF + daptomycin, vancomycin, meropenem or gentamycin combinations. Synergistic effects were generally obtained with BDSF + gentamicin combinations, followed by DSF + daptomycin against most S. aureus; while BDSF + gentamicin or ciprofloxacin, and DSF + vancomycin or meropenem were synergist against some S. epidermidis biofilms. Also, the antagonist effects were observed with BDSF + meropenem or ciprofloxacin against each MSSE and MSSA. It is estimated that these QS molecules, although it was strain dependent, generally enhanced the antibiotic activity, and would be a new and effective treatment strategy for biofilm control, either alone or as an antibiotic adjuvant.
Collapse
Affiliation(s)
- Merve Bilgin
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkiye
- Department of Pharmaceutical Microbiology, Istanbul Health & Technology University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Sibel Dosler
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Gulten Otuk
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| |
Collapse
|
21
|
D’Angelo C, Trecca M, Carpentieri A, Artini M, Selan L, Tutino ML, Papa R, Parrilli E. Cold-Azurin, a New Antibiofilm Protein Produced by the Antarctic Marine Bacterium Pseudomonas sp. TAE6080. Mar Drugs 2024; 22:61. [PMID: 38393032 PMCID: PMC10890351 DOI: 10.3390/md22020061] [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: 12/30/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Biofilm is accountable for nosocomial infections and chronic illness, making it a serious economic and public health problem. Staphylococcus epidermidis, thanks to its ability to form biofilm and colonize biomaterials, represents the most frequent causative agent involved in biofilm-associated infections of medical devices. Therefore, the research of new molecules able to interfere with S. epidermidis biofilm formation has a remarkable interest. In the present work, the attention was focused on Pseudomonas sp. TAE6080, an Antarctic marine bacterium able to produce and secrete an effective antibiofilm compound. The molecule responsible for this activity was purified by an activity-guided approach and identified by LC-MS/MS. Results indicated the active protein was a periplasmic protein similar to the Pseudomonas aeruginosa PAO1 azurin, named cold-azurin. The cold-azurin was recombinantly produced in E. coli and purified. The recombinant protein was able to impair S. epidermidis attachment to the polystyrene surface and effectively prevent biofilm formation.
Collapse
Affiliation(s)
- Caterina D’Angelo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (A.C.); (M.L.T.)
| | - Marika Trecca
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.T.); (M.A.); (L.S.); (R.P.)
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (A.C.); (M.L.T.)
| | - Marco Artini
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.T.); (M.A.); (L.S.); (R.P.)
| | - Laura Selan
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.T.); (M.A.); (L.S.); (R.P.)
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (A.C.); (M.L.T.)
| | - Rosanna Papa
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.T.); (M.A.); (L.S.); (R.P.)
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (A.C.); (M.L.T.)
| |
Collapse
|
22
|
Perez-Alba E, Flores-Treviño S, Villarreal-Salazar V, Bocanegra-Ibarias P, Vilchez-Cavazos F, Camacho-Ortiz A. Planktonic and biofilm states of Staphylococcus aureus isolated from bone and joint infections and the in vitro effect of orally available antibiotics. J Appl Microbiol 2023; 134:lxad258. [PMID: 37977856 DOI: 10.1093/jambio/lxad258] [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: 05/27/2023] [Revised: 10/02/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
AIMS To demonstrate the in vitro activity of orally available antibiotics against Staphylococcus aureus isolated from bone or orthopedic implant materials. The biofilm eradication of the combination of three antibiotics was also assessed. METHODS AND RESULTS Clinical isolates from orthopedic infection samples were collected, and S. aureus isolates were classified according to their biofilm production and composition. Almost all S. aureus isolates (n = 36, 97.3%) produced biofilm and the major biofilm components were polysaccharides. Antimicrobial susceptibility was determined in planktonic (minimal inhibitory concentration; MIC) and biofilm cells (minimal biofilm eradication concentration; MBEC) using the MBEC Calgary Device. Overall, the MBEC ranged higher than the MIC. When combined at borderline-susceptible concentrations, moxifloxacin-rifampin and doxycycline-rifampin were both able to eradicate biofilms in a third of the strains whereas the doxycycline-moxifloxacin combination proved ineffective at eradicating biofilm, inhibiting it only in three strains. CONCLUSIONS We propose rifampin in combination with moxifloxacin or doxycycline for the design of clinical trials of bone and/or orthopedic device infection without proper debridement or material retention.
Collapse
Affiliation(s)
- Eduardo Perez-Alba
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León., Av. Dr. José Eleuterio González S/N, Mitras Centro, 64460 Monterrey, N.L., Mexico
| | - Samantha Flores-Treviño
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León., Av. Dr. José Eleuterio González S/N, Mitras Centro, 64460 Monterrey, N.L., Mexico
| | - Verónica Villarreal-Salazar
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León., Pedro de Alba, Niños Héroes, Ciudad Universitaria, 66455 San Nicolás de los Garza, N.L., Mexico
| | - Paola Bocanegra-Ibarias
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León., Av. Dr. José Eleuterio González S/N, Mitras Centro, 64460 Monterrey, N.L., Mexico
| | - Félix Vilchez-Cavazos
- Servicio de Ortopedia y Traumatología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León., Av. Dr. José Eleuterio González S/N, Mitras Centro, 64460 Monterrey, N.L., Mexico
| | - Adrián Camacho-Ortiz
- Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León., Av. Dr. José Eleuterio González S/N, Mitras Centro, 64460 Monterrey, N.L., Mexico
- Departamento de Epidemiología Hospitalaria y Servicio de Infectología, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León., Av. Dr. José Eleuterio González S/N, Mitras Centro, 64460 Monterrey, N.L., Mexico
| |
Collapse
|
23
|
de Barros M, da Silva Lopes I, Moreira AJ, Dos Santos Oliveira Almeida R, Matiuzzi da Costa M, Mota RA, Nero LA, Scatamburlo Moreira MA. Multidrug Efflux System-mediated resistance in Staphylococcus aureus under a One Health approach. World J Microbiol Biotechnol 2023; 40:9. [PMID: 37938391 DOI: 10.1007/s11274-023-03793-z] [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: 12/13/2022] [Accepted: 10/05/2023] [Indexed: 11/09/2023]
Abstract
The aim of the study was to track the spread of antimicrobial resistance among the different sectors of One Health through the detection of Multidrug-Efflux-System in multidrug-resistant Staphylococcus aureus isolates. Multidrug-resistant (MDR) and methicillin-resistant (MRSA) S. aureus isolates were selected: 25 of human, one of animal and eight of food origin. The efflux system genes norA, norB, norC, LmrS, tet38 and msrA were screened by PCR. The activity of the efflux systems was determined by the minimum inhibitory concentration (MIC) of tetracycline and ciprofloxacin in the presence and absence of CCCP and in the quantification of ethidium bromide efflux. Furthermore, biofilm formation was determined in the presence and absence of the CCCP. The molecular epidemiology of the isolates was traced with the aid of PFGE. The gene norC was the most prevalent, detected in all isolates and msrA was the least prevalent, detected in only two isolates from humans. There was no difference in the MICs of tetracycline and ciprofloxacin in the presence of CCCP, but 55.9% of isolates showed ethidium bromide efflux. The presence of CCCP decreased the biofilm formation. Regarding the molecular epidemiology, in three clusters was a mixture of the isolates from different origins. Therefore, S. aureus MDR with active multidrug efflux systems are circulating between One Health domains and it is necessary to consider strategies to decrease this circulation in order to prevent the dissemination of resistance mediated by MES.
Collapse
Affiliation(s)
- Mariana de Barros
- Department of Veterinary, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | | - Ana Júlia Moreira
- Department of Veterinary, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | | | | | | - Luis Augusto Nero
- Department of Veterinary, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | |
Collapse
|
24
|
Casillo A, D’Angelo C, Imbimbo P, Monti DM, Parrilli E, Lanzetta R, Gomez d’Ayala G, Mallardo S, Corsaro MM, Duraccio D. Aqueous Extracts from Hemp Seeds as a New Weapon against Staphylococcus epidermidis Biofilms. Int J Mol Sci 2023; 24:16026. [PMID: 38003214 PMCID: PMC10671263 DOI: 10.3390/ijms242216026] [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: 10/07/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
This study investigated the antibiofilm activity of water-soluble extracts obtained under different pH conditions from Cannabis sativa seeds and from previously defatted seeds. The chemical composition of the extracts, determined through GC-MS and NMR, revealed complex mixtures of fatty acids, monosaccharides, amino acids and glycerol in ratios depending on extraction pH. In particular, the extract obtained at pH 7 from defatted seeds (Ex7d) contained a larger variety of sugars compared to the others. Saturated and unsaturated fatty acids were found in all of the analysed extracts, but linoleic acid (C18:2) was detected only in the extracts obtained at pH 7 and pH 10. The extracts did not show cytotoxicity to HaCaT cells and significantly inhibited the formation of Staphylococcus epidermidis biofilms. The exception was the extract obtained at pH 10, which appeared to be less active. Ex7d showed the highest antibiofilm activity, i.e., around 90%. Ex7d was further fractionated by HPLC, and the antibiofilm activity of all fractions was evaluated. The 2D-NMR analysis highlighted that the most active fraction was largely composed of glycerolipids. This evidence suggested that these molecules are probably responsible for the observed antibiofilm effect but does not exclude a possible synergistic contribution by the other components.
Collapse
Affiliation(s)
- Angela Casillo
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Caterina D’Angelo
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Paola Imbimbo
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Daria Maria Monti
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Rosa Lanzetta
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Giovanna Gomez d’Ayala
- Institute of Polymers, Composites and Biomaterials (IPCB)-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
| | - Salvatore Mallardo
- Institute of Polymers, Composites and Biomaterials (IPCB)-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
| | - Maria Michela Corsaro
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 21, 80126 Napoli, Italy; (A.C.); (C.D.); (P.I.); (D.M.M.); (E.P.); (R.L.); (M.M.C.)
| | - Donatella Duraccio
- Institute of Sciences and Technologies for Sustainable Energy and Mobility (STEMS)-CNR, Strada Delle Cacce 73, 10135 Torino, Italy;
| |
Collapse
|
25
|
Yang S, Li X, Cang W, Mu D, Ji S, An Y, Wu R, Wu J. Biofilm tolerance, resistance and infections increasing threat of public health. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:233-247. [PMID: 37933277 PMCID: PMC10625689 DOI: 10.15698/mic2023.11.807] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 11/08/2023]
Abstract
Microbial biofilms can cause chronic infection. In the clinical setting, the biofilm-related infections usually persist and reoccur; the main reason is the increased antibiotic resistance of biofilms. Traditional antibiotic therapy is not effective and might increase the threat of antibiotic resistance to public health. Therefore, it is urgent to study the tolerance and resistance mechanism of biofilms to antibiotics and find effective therapies for biofilm-related infections. The tolerance mechanism and host reaction of biofilm to antibiotics are reviewed, and bacterial biofilm related diseases formed by human pathogens are discussed thoroughly. The review also explored the role of biofilms in the development of bacterial resistance mechanisms and proposed therapeutic intervention strategies for biofilm related diseases.
Collapse
Affiliation(s)
- Shanshan Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Xinfei Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
| | - Weihe Cang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
| | - Delun Mu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Shuaiqi Ji
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Yuejia An
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| |
Collapse
|
26
|
Zhou X, Wells MJ, Gordon VD. Incorporation of collagen into Pseudomonas aeruginosa and Staphylococcus aureus biofilms impedes phagocytosis by neutrophils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.25.564018. [PMID: 37961328 PMCID: PMC10634824 DOI: 10.1101/2023.10.25.564018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Biofilms are communities of microbes embedded in a matrix of extracellular polymeric substances (EPS). Matrix components can be produced by biofilm organisms and can also originate from the environment and then be incorporated into the biofilm. For example, we have recently shown that collagen, a host-produced protein that is abundant in many different infection sites, can be taken up into the biofilm matrix, altering biofilm mechanics. The biofilm matrix protects bacteria from clearance by the immune system, and some of that protection likely arises from the mechanical properties of the biofilm. Pseudomonas aeruginosa and Staphylococcus aureus are common human pathogens notable for forming biofilm infections in anatomical sites rich in collagen. Here, we show that the incorporation of Type I collagen into P. aeruginosa and S. aureus biofilms significantly hinders phagocytosis of biofilm bacteria by human neutrophils. However, enzymatic treatment with collagenase, which breaks down collagen, can partly or entirely negate the protective effect of collagen and restore the ability of neutrophils to engulf biofilm bacteria. From these findings, we suggest that enzymatic degradation of host materials may be a potential way to compromise biofilm infections and enhance the efficacy of the host immune response without promoting antibiotic resistance. Such an approach might be beneficial both in cases where the infecting species is known and also in cases wherein biofilm components are not readily known, such as multispecies infections or infections by unknown species.
Collapse
Affiliation(s)
- Xuening Zhou
- Center for Nonlinear Dynamics, The University of Texas at Austin, 2515 Speedway, Stop C1610, Austin, Texas 78712-11993, USA
- Interdisciplinary Life Sciences Graduate Program, Norman Hackerman Building, 100 East 24th St., NHB 4500, Austin, Texas 78712, USA
| | - Marilyn J Wells
- Center for Nonlinear Dynamics, The University of Texas at Austin, 2515 Speedway, Stop C1610, Austin, Texas 78712-11993, USA
- Department of Physics, The University of Texas at Austin, 2515 Speedway, C1600, Austin, Texas 78712-1192, USA
| | - Vernita D Gordon
- Center for Nonlinear Dynamics, The University of Texas at Austin, 2515 Speedway, Stop C1610, Austin, Texas 78712-11993, USA
- Interdisciplinary Life Sciences Graduate Program, Norman Hackerman Building, 100 East 24th St., NHB 4500, Austin, Texas 78712, USA
- Department of Physics, The University of Texas at Austin, 2515 Speedway, C1600, Austin, Texas 78712-1192, USA
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Neural Molecular Science Building, 2506 Speedway, Stop A5000, Austin, Texas 78712, USA
| |
Collapse
|
27
|
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: 2.0] [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
|
28
|
Caciandone-Cringureanu M, Anghel AG, Anghel I. Biofilms on Voice Prosthesis - Challenges and Therapeutic Insights. MAEDICA 2023; 18:498-503. [PMID: 38023755 PMCID: PMC10674123 DOI: 10.26574/maedica.2023.18.3.498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
After a total laryngectomy, voice rehabilitation is essential for a patient to successfully reintegrate into society. The implantation of a voice prosthesis (VP) is the gold standard to achieve this goal. Thus, the primary disadvantage of using VP is the fluid blockage and degradation caused by biofilm colonization, which requires frequent replacements, associated with a poor quality of life for the patient. Many scientists have centered their research on coming up with novel and efficient ways to combat polymicrobial biofilms, both in terms of preventing microbial adhesion and rupturing established biofilms in order to overcome this limitation. This paper aims to present the current state of the art regarding biofilm formation on VPs and composition of VPs, and to review the current anti-biofilm strategies that have proven to be successful, as well as pointing possible novel perspectives of improvement.
Collapse
Affiliation(s)
| | - Alina-Georgiana Anghel
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- ENT Department, Saint Mary Clinical Hospital Bucharest, Romania
| | - Ion Anghel
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Dr. Carol Davila" Central Military Emergency University Hospital, 010825 Bucharest, Romania
| |
Collapse
|
29
|
Ruiz-Romero RA, Vargas-Bello-Pérez E. Non-aureus staphylococci and mammaliicocci as a cause of mastitis in domestic ruminants: current knowledge, advances, biomedical applications, and future perspectives - a systematic review. Vet Res Commun 2023; 47:1067-1084. [PMID: 36964436 PMCID: PMC10038778 DOI: 10.1007/s11259-023-10090-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/24/2023] [Indexed: 03/26/2023]
Abstract
Non-aureus staphylococci and mammaliicocci (NASM) are one of the most common causes of subclinical mastitis in dairy animals and the extent of damage by intramammary infections (IMI) caused by NASM is still under debate. The different effects of NASM on the mammary gland may be associated with differences between bacterial species. NASM are normal and abundant colonizers of humans and animals and become pathogenic only in certain situations. The veterinary interest in NASM has been intense for the last 25 years, due to the strongly increasing rate of opportunistic infections. Therefore, the objective of this review is to provide a general background of the NASM as a cause of mastitis and the most recent advances that exist to prevent and fight the biofilm formation of this group of bacteria, introduce new biomedical applications that could be used in dairy herds to reduce the risk of chronic and recurrent infections, potentially responsible for economic losses due to reduced milk production and quality. Effective treatment of biofilm infection requires a dual approach through a combination of antibiofilm and antimicrobial agents. Even though research on the development of biofilms is mainly focused on human medicine, this technology must be developed at the same time in veterinary medicine, especially in the dairy industry where IMI are extremely common.
Collapse
Affiliation(s)
- Rocio Angélica Ruiz-Romero
- Departamento de Medicina y Zootecnia de Rumiantes, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Av. Universidad 3000, Ciudad de México, 04510, México.
| | - Einar Vargas-Bello-Pérez
- School of Agriculture, Policy and Development, University of Reading, New Agriculture Building, Earley Gate, Whiteknights Road, PO Box 237, Reading, Berkshire, RG6 6EU, UK.
| |
Collapse
|
30
|
Giugliano R, Della Sala G, Buonocore C, Zannella C, Tedesco P, Palma Esposito F, Ragozzino C, Chianese A, Morone MV, Mazzella V, Núñez-Pons L, Folliero V, Franci G, De Filippis A, Galdiero M, de Pascale D. New Imidazolium Alkaloids with Broad Spectrum of Action from the Marine Bacterium Shewanella aquimarina. Pharmaceutics 2023; 15:2139. [PMID: 37631353 PMCID: PMC10458398 DOI: 10.3390/pharmaceutics15082139] [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: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The continuous outbreak of drug-resistant bacterial and viral infections imposes the need to search for new drug candidates. Natural products from marine bacteria still inspire the design of pharmaceuticals. Indeed, marine bacteria have unique metabolic flexibility to inhabit each ecological niche, thus expanding their biosynthetic ability to assemble unprecedented molecules. The One-Strain-Many-Compounds approach and tandem mass spectrometry allowed the discovery of a Shewanella aquimarina strain as a source of novel imidazolium alkaloids via molecular networking. The alkaloid mixture was shown to exert bioactivities such as: (a) antibacterial activity against antibiotic-resistant Staphylococcus aureus clinical isolates at 100 µg/mL, (b) synergistic effects with tigecycline and linezolid, (c) restoration of MRSA sensitivity to fosfomycin, and (d) interference with the biofilm formation of S. aureus 6538 and MRSA. Moreover, the mixture showed antiviral activity against viruses with and without envelopes. Indeed, it inhibited the entry of coronavirus HcoV-229E and herpes simplex viruses into human cells and inactivated poliovirus PV-1 in post-infection assay at 200 µg/mL. Finally, at the same concentration, the fraction showed anthelminthic activity against Caenorhabditis elegans, causing 99% mortality after 48 h. The broad-spectrum activities of these compounds are partially due to their biosurfactant behavior and make them promising candidates for breaking down drug-resistant infectious diseases.
Collapse
Affiliation(s)
- Rosa Giugliano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.G.); (C.Z.); (A.C.); (M.V.M.); (A.D.F.)
| | - Gerardo Della Sala
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Acton, 55, 80133 Naples, Italy; (G.D.S.); (C.B.); (P.T.); (F.P.E.); (C.R.)
| | - Carmine Buonocore
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Acton, 55, 80133 Naples, Italy; (G.D.S.); (C.B.); (P.T.); (F.P.E.); (C.R.)
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.G.); (C.Z.); (A.C.); (M.V.M.); (A.D.F.)
| | - Pietro Tedesco
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Acton, 55, 80133 Naples, Italy; (G.D.S.); (C.B.); (P.T.); (F.P.E.); (C.R.)
| | - Fortunato Palma Esposito
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Acton, 55, 80133 Naples, Italy; (G.D.S.); (C.B.); (P.T.); (F.P.E.); (C.R.)
| | - Costanza Ragozzino
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Acton, 55, 80133 Naples, Italy; (G.D.S.); (C.B.); (P.T.); (F.P.E.); (C.R.)
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.G.); (C.Z.); (A.C.); (M.V.M.); (A.D.F.)
| | - Maria Vittoria Morone
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.G.); (C.Z.); (A.C.); (M.V.M.); (A.D.F.)
| | - Valerio Mazzella
- Department of Integrative Marine Ecology (EMI), Stazione Zoologica Anton Dohrn, Ischia Marine Centre, Ischia, 80077 Naples, Italy;
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy;
| | - Laura Núñez-Pons
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy;
- Department of Integrative Marine Ecology (EMI), Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
| | - Veronica Folliero
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (G.F.)
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (G.F.)
| | - Anna De Filippis
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.G.); (C.Z.); (A.C.); (M.V.M.); (A.D.F.)
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.G.); (C.Z.); (A.C.); (M.V.M.); (A.D.F.)
| | - Donatella de Pascale
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Acton, 55, 80133 Naples, Italy; (G.D.S.); (C.B.); (P.T.); (F.P.E.); (C.R.)
| |
Collapse
|
31
|
Hussain SA, Walters S, Ahluwalia AK, Trompeter A. Fracture-related infections. Br J Hosp Med (Lond) 2023; 84:1-10. [PMID: 37646543 DOI: 10.12968/hmed.2022.0545] [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: 09/01/2023]
Abstract
Fracture-related infection is a serious complication which can occur following musculoskeletal injury and is associated with significant morbidity. These complications can be challenging to recognise, and experts have provided a clearer definition of fracture-related infection to help with the diagnosis and detection of these infections. This system includes clinical, radiological and laboratory-based diagnostic features which are either confirmatory or suggestive of fracture-related infection. Treatment requires a multifaceted approach with multidisciplinary involvement, and generally a combination of surgical techniques and prolonged antibiotics, the timing and choice of which should be optimised. This article provides an evidence-based review of the British Orthopaedic Association Standards for Trauma for the diagnosis and management of fracture-related infections.
Collapse
Affiliation(s)
| | - Samuel Walters
- Orthopaedic Trauma/Limb Reconstruction Unit, St George's University Hospital, London, UK
| | - Aashish K Ahluwalia
- Department of Trauma and Orthopaedic Surgery, University College Hospital, London, UK
| | - Alex Trompeter
- Orthopaedic Trauma/Limb Reconstruction Unit, St George's University Hospital, London, UK
| |
Collapse
|
32
|
Aldayel MF. Enhancement of the Bioactive Compound Content and Antibacterial Activities in Curcuma Longa Using Zinc Oxide Nanoparticles. Molecules 2023; 28:4935. [PMID: 37446597 DOI: 10.3390/molecules28134935] [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: 05/28/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Incorporating nanoparticles into plant cultivation has been shown to improve growth parameters and alter the bioactive component compositions of many plant species, including Curcumin longa. The aim of the current study was to investigate the effects of foliar application of zinc oxide nanoparticles on the content of bioactive compounds and their antibacterial activities against potential bacterial pathogens. To this end, C. longa leaves were treated with different doses of ZnO NPs to see how this affected their bioactive component composition. The effect of different doses of ZnO NPs on the accumulation of bisdemethoxycurcumin, demethoxycurcumin, and curcumin in ethanolic extracts of C. longa rhizomes was evaluated using high-performance liquid chromatography (HPLC). When compared to the control treatment, foliar spraying with (5 and 40 mgL-1) of ZnO NPs increased bisdemethoxycurcumin, demethoxycurcumin, and curcumin levels approximately (2.69 and 2.84)-, (2.61 and 3.22)-, and (2.90 and 3.45)-fold, respectively. We then checked whether the ethanolic extracts produced from the plantlets changed in terms of their phytochemical makeup and antibacterial properties. Furthermore, the results revealed that C. long-ZnO NPs displayed antibacterial activity against the tested S. aureus and P. aeruginosa bacterium strains, but had a few effect against E. coli. The MIC for P. aeruginosa was 100 g/mL. The time-kill studies also revealed that ZnO NPs at 4 MIC killed P. aeruginosa, Actinobacteria baumannii, and Bacillus sp. after 2 h, while S. aureus did not grow when treated with 4 × MIC of the extract for 6 h. The strongest antibacterial activity was seen in the extract from plantlets grown without nanoparticles for P. aeruginosa, whereas it was seen in the extract from plantlets grown in the presence of 5 mg/L ZnO NPs for E. coli, S. aureus, and P. aeruginosa. These findings show that ZnO NPs are powerful enhancers of bioactive compound production in C. longa, a trait that can be used to combat antibiotic resistance in pathogenic bacterial species.
Collapse
Affiliation(s)
- Munirah F Aldayel
- Department of Biological Sciences, College of Science, King Faisal University, AlAhsa 31982, Saudi Arabia
| |
Collapse
|
33
|
Zhao A, Sun J, Liu Y. Understanding bacterial biofilms: From definition to treatment strategies. Front Cell Infect Microbiol 2023; 13:1137947. [PMID: 37091673 PMCID: PMC10117668 DOI: 10.3389/fcimb.2023.1137947] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Bacterial biofilms are complex microbial communities encased in extracellular polymeric substances. Their formation is a multi-step process. Biofilms are a significant problem in treating bacterial infections and are one of the main reasons for the persistence of infections. They can exhibit increased resistance to classical antibiotics and cause disease through device-related and non-device (tissue) -associated infections, posing a severe threat to global health issues. Therefore, early detection and search for new and alternative treatments are essential for treating and suppressing biofilm-associated infections. In this paper, we systematically reviewed the formation of bacterial biofilms, associated infections, detection methods, and potential treatment strategies, aiming to provide researchers with the latest progress in the detection and treatment of bacterial biofilms.
Collapse
Affiliation(s)
- Ailing Zhao
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Jiazheng Sun
- Department of Vasculocardiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yipin Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
- *Correspondence: Yipin Liu,
| |
Collapse
|
34
|
Li M, Yu J, Guo G, Shen H. Interactions between Macrophages and Biofilm during Staphylococcus aureus-Associated Implant Infection: Difficulties and Solutions. J Innate Immun 2023; 15:499-515. [PMID: 37011602 PMCID: PMC10315156 DOI: 10.1159/000530385] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/16/2023] [Indexed: 04/05/2023] Open
Abstract
Staphylococcus aureus (S. aureus) biofilm is the major cause of failure of implant infection treatment that results in heavy social and economic burden on individuals, families, and communities. Planktonic S. aureus attaches to medical implant surfaces where it proliferates and is wrapped by extracellular polymeric substances, forming a solid and complex biofilm. This provides a stable environment for bacterial growth, infection maintenance, and diffusion and protects the bacteria from antimicrobial agents and the immune system of the host. Macrophages are an important component of the innate immune system and resist pathogen invasion and infection through phagocytosis, antigen presentation, and cytokine secretion. The persistence, spread, or clearance of infection is determined by interplay between macrophages and S. aureus in the implant infection microenvironment. In this review, we discuss the interactions between S. aureus biofilm and macrophages, including the effects of biofilm-related bacteria on the macrophage immune response, roles of myeloid-derived suppressor cells during biofilm infection, regulation of immune cell metabolic patterns by the biofilm environment, and immune evasion strategies adopted by the biofilm against macrophages. Finally, we summarize the current methods that support macrophage-mediated removal of biofilms and emphasize the importance of considering multi-dimensions and factors related to implant-associated infection such as immunity, metabolism, the host, and the pathogen when developing new treatments.
Collapse
Affiliation(s)
- Mingzhang Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinlong Yu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Geyong Guo
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Shen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Zhao D, Li X, Xu M, Jiao Y, Liu H, Xiao X, Zhao H. Preparations of antibacterial yellow-green-fluorescent carbon dots and carbon dots-lysozyme complex and their applications in bacterial imaging and bacteria/biofilm inhibition/clearance. Int J Biol Macromol 2023; 231:123303. [PMID: 36657551 DOI: 10.1016/j.ijbiomac.2023.123303] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
The preparation of functional long-wavelength-emitting nanomaterials and the researches on their applications in antibacterial and antibiofilm fields have important significance. This paper reports the preparation of yellow-green-fluorescent and high- quantum yield carbon dots (4-ACDs) with 4-aminosalicylic acid and polyethylene imine as raw materials through one-step route, and the impacts of raw material structure and the reaction conditions upon the optical properties of the products have been investigated. 4-ACDs exhibit excellent broad-spectrum antibacterial activity, and their good biocompatibility ensures them as ideal fluorescent nano-probe for cell imaging. However, 4-ACDs could not effectively eliminate the biofilm of Staphylococcus aureus (S. aureus). CDs-LZM complex was prepared through the coupling between 4-ACDs and lysozyme (LZM) and the complex showed strong antibacterial activity against Gram-positive bacteria, particularly with MIC against S. aureus at 5 μg mL-1. Besides, CDs-LZM showed excellent ability against the biofilm of S. aureus. At the concentration of 60 μg mL-1, its inhibition rate against the growth of biofilm was 86 %, and elimination rate against biofilm reached 76 %. CDs-LZM exhibited obvious antibiofilm ability through removing extracellular matrix of biofilm, greatly reducing the thickness of biofilm under confocal microscopy. The application of novel long-wavelength-emitting nanomaterial in eliminating pathogenic bacteria is of great significance.
Collapse
Affiliation(s)
- Dan Zhao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China.
| | - Xiaoyun Li
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China
| | - Mengyu Xu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China
| | - Yan Jiao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China
| | - Huan Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China
| | - Xincai Xiao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China
| | - Haiyan Zhao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, PR China; National Demonstration Center for Experimental Ethnopharmacology Education (South-Central Minzu University), Wuhan 430065, PR China
| |
Collapse
|
36
|
França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics (Basel) 2023; 12:antibiotics12030554. [PMID: 36978421 PMCID: PMC10044083 DOI: 10.3390/antibiotics12030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.
Collapse
Affiliation(s)
- Angela França
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
| |
Collapse
|
37
|
Sekar A, Lekkala S, Oral E. A Novel Method to Determine the Longitudinal Antibacterial Activity of Drug-Eluting Materials. J Vis Exp 2023:10.3791/64641. [PMID: 36939249 PMCID: PMC10859037 DOI: 10.3791/64641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Ultrahigh molecular weight polyethylene (UHMWPE) is widely used in total joint arthroplasties as a load-bearing surface. Periprosthetic joint infections, the majority of which occur shortly after joint replacement, constitute almost 25% of total knee revision surgeries, and the complete eradication of bacterial infection poses a major challenge. A promising way to tackle this problem is to ensure the local sustained delivery of antibiotic concentrations sufficient to inhibit the bacteria to support routine systemic antibiotic prophylaxis. There is increased research into the development of efficient local drug delivery devices. Although established antibacterial testing methods for drugs can be used to test the antibacterial efficacy of drug-eluting materials, they are lacking in terms of providing real-time and longitudinal antibacterial efficacy data that can be correlated to the elution profiles of antibiotics from these devices. Here, we report a direct and versatile methodology to determine the antibacterial efficacy of antibiotic-eluting UHMWPE implants. This methodology can be used as a platform to avoid bacterial culture at each time point of a lengthy experiment and can also be adapted to other local drug delivery devices.
Collapse
Affiliation(s)
- Amita Sekar
- Harris Orthopaedics Laboratory, Massachusetts General Hospital; Department of Orthopaedic Surgery, Harvard Medical School
| | - Sashank Lekkala
- Harris Orthopaedics Laboratory, Massachusetts General Hospital
| | - Ebru Oral
- Harris Orthopaedics Laboratory, Massachusetts General Hospital; Department of Orthopaedic Surgery, Harvard Medical School;
| |
Collapse
|
38
|
Aleksandrova YI, Shurpik DN, Nazmutdinova VA, Mostovaya OA, Subakaeva EV, Sokolova EA, Zelenikhin PV, Stoikov II. Toward Pathogenic Biofilm Suppressors: Synthesis of Amino Derivatives of Pillar[5]arene and Supramolecular Assembly with DNA. Pharmaceutics 2023; 15:pharmaceutics15020476. [PMID: 36839796 PMCID: PMC9966598 DOI: 10.3390/pharmaceutics15020476] [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: 12/25/2022] [Revised: 01/28/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
New amino derivatives of pillar[5]arene were obtained in three stages with good yields. It was shown that pillar[5]arene containing thiaether and tertiary amino groups formed supramolecular complexes with low molecular weight model DNA. Pillar[5]arene formed complexes with a DNA nucleotide pair at a ratio of 1:2 (macrocycle/DNA base pairs), as demonstrated by UV-visible and fluorescence spectroscopy. The association constants of pillar[5]arene with DNA were lgKass1:1 = 2.38 and lgKass1:2 = 5.07, accordingly. By using dynamic light scattering and transmission electron microscopy, it was established that the interaction of pillar[5]arene containing thiaether and tertiary amino groups (concentration of 10-5 M) with a model nucleic acid led to the formation of stable nanosized macrocycle/DNA associates with an average particle size of 220 nm. It was shown that the obtained compounds did not exhibit a pronounced toxicity toward human adenocarcinoma cells (A549) and bovine lung epithelial cells (LECs). The hypothesis about a possible usage of the synthesized macrocycle for the aggregation of extracellular bacterial DNA in a biofilm matrix was confirmed by the example of St. Aureus. It was found that pillar[5]arene at a concentration of 10-5 M was able to reduce the thickness of the St. Aureus biofilm by 15%.
Collapse
Affiliation(s)
- Yulia I. Aleksandrova
- A.M. Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
| | - Dmitriy N. Shurpik
- A.M. Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
- Correspondence: (D.N.S.); (I.I.S.); Tel.: +7-843-233-7241 (I.I.S.)
| | | | - Olga A. Mostovaya
- A.M. Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
| | - Evgenia V. Subakaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
| | - Evgenia A. Sokolova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
| | - Pavel V. Zelenikhin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
| | - Ivan I. Stoikov
- A.M. Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya, 18, 420008 Kazan, Russia
- Correspondence: (D.N.S.); (I.I.S.); Tel.: +7-843-233-7241 (I.I.S.)
| |
Collapse
|
39
|
Sulaiman R, Trizna E, Kolesnikova A, Khabibrakhmanova A, Kurbangalieva A, Bogachev M, Kayumov A. Antimicrobial and Biofilm-Preventing Activity of l-Borneol Possessing 2(5H)-Furanone Derivative F131 against S. aureus—C. albicans Mixed Cultures. Pathogens 2022; 12:pathogens12010026. [PMID: 36678375 PMCID: PMC9866062 DOI: 10.3390/pathogens12010026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Candida albicans and Staphylococcus aureus are human pathogens that are able to form mixed biofilms on the surface of mucous membranes, implants and catheters. In biofilms, these pathogens have increased resistance to antimicrobials, leading to extreme difficulties in the treatment of mixed infections. The growing frequency of mixed infections caused by S. aureus and C. albicans requires either the development of new antimicrobials or the proposal of alternative approaches to increase the efficiency of conventional ones. Here, we show the antimicrobial, biofilm-preventing and biofilm-eradicating activity of 2(5H)-furanone derivative F131, containing an l-borneol fragment against S. aureus-C. albicans mixed biofilms. Furanone F131 is also capable of inhibiting the formation of monospecies and mixed biofilms by S. aureus and C. albicans. The minimal biofilm-prevention concentration (MBPC) of this compound was 8-16 μg/mL for S. aureus and C. albicans mono- and two-species biofilms. While the compound demonstrates slightly lower activity compared to conventional antimicrobials (gentamicin, amikacin, fluconazole, terbinafine and benzalkonium chloride), F131 also increases the antimicrobial activity of fluconazole-gentamicin and benzalkonium chloride against mixed biofilms of S. aureus-C. albicans, thus reducing MBPC of fluconazole-gentamicin by 4-16 times and benzalkonium chloride twofold. F131 does not affect the transcription of the MDR1, CDR1 and CDR2 genes, thus suggesting a low risk of micromycete resistance to this compound. Altogether, combined use of antibiotics with a F131 could be a promising option to reduce the concentration of fluconazole used in antiseptic compositions and reduce the toxic effect of benzalkonium chloride and gentamicin. This makes them an attractive starting point for the development of alternative antimicrobials for the treatment of skin infections caused by S. aureus-C. albicans mixed biofilms.
Collapse
Affiliation(s)
- Rand Sulaiman
- Laboratory of Molecular Genetics of Microorganisms, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Elena Trizna
- Laboratory of Molecular Genetics of Microorganisms, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Alena Kolesnikova
- Laboratory of Molecular Genetics of Microorganisms, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Alsu Khabibrakhmanova
- Biofunctional Chemistry Laboratory, Alexander Butlerov Institute of Chemistry, Kazan Federal University, 420008 Kazan, Russia
| | - Almira Kurbangalieva
- Biofunctional Chemistry Laboratory, Alexander Butlerov Institute of Chemistry, Kazan Federal University, 420008 Kazan, Russia
| | - Mikhail Bogachev
- Biomedical Engineering Research Centre, St. Petersburg Electrotechnical University, 197022 St. Petersburg, Russia
| | - Airat Kayumov
- Laboratory of Molecular Genetics of Microorganisms, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Correspondence:
| |
Collapse
|
40
|
Proof-of-Concept Standardized Approach Using a Single-Disk Method Analogous to Antibiotic Disk Diffusion Assays for Routine Phage Susceptibility Testing in Diagnostic Laboratories. Appl Environ Microbiol 2022; 88:e0030922. [PMID: 36416554 PMCID: PMC9746313 DOI: 10.1128/aem.00309-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Application of bacteriophages is increasingly being implemented in clinical therapies. Prior susceptibility testing should be regarded as mandatory, but standards are lacking. The objective of this research was to develop a highly standardized methodology to facilitate phage susceptibility testing (PST) in clinical microbiology routine laboratories. Therefore, EUCAST methods established for single disk-based antibiotic susceptibility testing (AST) were adapted. In a first step, basic parameters were evaluated using well-studied Escherichia phage T4-Escherichia coli combinations. In addition, test results were compared to those from conventional spot test and efficiency of plating (EOP) approaches. In a second step, the applicability of the methodology and the most promising test parameters were demonstrated for five other frequently isolated clinical bacterial species and their corresponding phages. At present, the method predominantly leads to qualitative rather than quantitative results. This disk-based approach provides a standardized, easy-to-handle, reproducible and reliable PST protocol by relying on well-established routine procedures in diagnostic laboratories. IMPORTANCE Application of bacteriophages in clinical therapies is attractive due to increasing rates of isolation of multidrug-resistant bacteria worldwide. As the phage effect is highly specific, prior susceptibility testing of target bacteria is mandatory. Of note, established standards are lacking. In this research, we adapted the single-disk method for antibiotic susceptibility testing to phage susceptibility testing (PST) in order to provide a standardized, easy-to-handle, reproducible, and reliable PST protocol for application in diagnostic routine laboratories.
Collapse
|
41
|
Alvira-Arill GR, Herrera OR, Tsang CCS, Wang J, Peters BM, Stultz JS. Comparison of catheter-related bloodstream infection rates in pediatric patients receiving parenteral nutrition with soybean oil-based intravenous fat emulsion versus a mixed oil fat emulsion. Pharmacotherapy 2022; 42:898-904. [PMID: 36349792 PMCID: PMC9742122 DOI: 10.1002/phar.2740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
Abstract
STUDY OBJECTIVE To compare rates of catheter-related bloodstream infections (CR-BSI) in pediatric patients who received parenteral nutrition (PN) with either soybean oil-based intravenous fat emulsion (SO-IFE) or mixed oil-IFE (MO-IFE). We hypothesized that the use of MO-IFE would be independently associated with reduced infection rates compared with SO-IFE. DESIGN Retrospective cohort study. SETTING Tertiary referral children's hospital and its associated gastrointestinal rehabilitation clinic (01 January, 2015-31 July, 2019). PATIENTS Days of IFE exposure were counted for patients aged <18 years on IFE initiated during the review period, who had a central venous catheter (CVC) placed for PN administration, received IFE at least three times weekly, and for at least 7 days. MEASUREMENTS The primary outcome included total and categorical CR-BSI rates expressed as the average with standard error (SE) number of infections per 1000 fat emulsion days. The following categories were specified: Candida albicans, non-albicans Candida spp., coagulase-negative Staphylococcus (CoNS), Enterobacterales, methicillin-resistant S. aureus, methicillin-susceptible S. aureus, and Pseudomonadales. Average infection rate comparisons were quantified as incidence rate ratios (IRR) using generalized linear mixed modeling with a Poisson distribution. MAIN RESULTS Seven hundred and forty-three SO-IFE and 450 MO-IFE exposures were reviewed from 1131 patients, totaling 37,599 and 19,796 days of therapy, respectively. From those found significantly different, the average rate of infections with CoNS was 3.58 (SE 0.5)/1000 days of SO-IFE and 1.39 (SE 0.45)/1000 days of MO-IFE (IRR [95% confidence interval, CI]: 0.27 [0.16-0.46]; p < 0.01). Total average rates of infection were 7.33 (SE 0.76)/1000 days of SO-IFE and 4.52 (SE 0.75)/1000 days of MO-IFE (IRR [95% CI]: 0.60 [0.44-0.81]; p < 0.01). Other factors associated with higher infection rates include female gender, neonatal age, and inpatient-only IFE exposure. CONCLUSIONS Receipt of MO-IFE was associated with lower rates of CoNS and total CR-BSIs compared with SO-IFE in pediatric patients. These findings could have major implications on IFE selection for pediatric patients receiving PN.
Collapse
Affiliation(s)
- Gustavo R. Alvira-Arill
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Pharmacy, Le Bonheur Children’s Hospital, Memphis, Tennessee, USA
| | - Oscar R. Herrera
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Pharmacy, Le Bonheur Children’s Hospital, Memphis, Tennessee, USA
| | - Chi Chun Steve Tsang
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Junling Wang
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Brian M. Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jeremy S. Stultz
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Pharmacy, Le Bonheur Children’s Hospital, Memphis, Tennessee, USA
| |
Collapse
|
42
|
D’Angelo C, Casillo A, Melchiorre C, Lauro C, Corsaro MM, Carpentieri A, Tutino ML, Parrilli E. CATASAN Is a New Anti-Biofilm Agent Produced by the Marine Antarctic Bacterium Psychrobacter sp. TAE2020. Mar Drugs 2022; 20:md20120747. [PMID: 36547894 PMCID: PMC9785100 DOI: 10.3390/md20120747] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
The development of new approaches to prevent microbial surface adhesion and biofilm formation is an emerging need following the growing understanding of the impact of biofilm-related infections on human health. Staphylococcus epidermidis, with its ability to form biofilm and colonize biomaterials, represents the most frequent causative agent involved in infections of medical devices. In the research of new anti-biofilm agents against S. epidermidis biofilm, Antarctic marine bacteria represent an untapped reservoir of biodiversity. In the present study, the attention was focused on Psychrobacter sp. TAE2020, an Antarctic marine bacterium that produces molecules able to impair the initial attachment of S. epidermidis strains to the polystyrene surface. The setup of suitable purification protocols allowed the identification by NMR spectroscopy and LC-MS/MS analysis of a protein-polysaccharide complex named CATASAN. This complex proved to be a very effective anti-biofilm agent. Indeed, it not only interferes with cell surface attachment, but also prevents biofilm formation and affects the mature biofilm matrix structure of S. epidermidis. Moreover, CATASAN is endowed with a good emulsification activity in a wide range of pH and temperature. Therefore, its use can be easily extended to different biotechnological applications.
Collapse
Affiliation(s)
- Caterina D’Angelo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
| | - Angela Casillo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
| | - Chiara Melchiorre
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
| | - Concetta Lauro
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
- Istituto Nazionale Biostrutture e Biosistemi—I.N.B.B., Viale Medaglie d’Oro, 305-00136 Rome, Italy
| | - Maria Michela Corsaro
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
- Correspondence: ; Tel.: +39-081674003; Fax: +39-081674113
| |
Collapse
|
43
|
Gopalakrishnan S, Gupta A, Makabenta JMV, Park J, Amante JJ, Chattopadhyay AN, Matuwana D, Kearney CJ, Rotello VM. Ultrasound-Enhanced Antibacterial Activity of Polymeric Nanoparticles for Eradicating Bacterial Biofilms. Adv Healthc Mater 2022; 11:e2201060. [PMID: 36049222 PMCID: PMC9633556 DOI: 10.1002/adhm.202201060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/26/2022] [Indexed: 01/28/2023]
Abstract
Bacterial biofilms are a major healthcare concern resulting in refractory conditions such as chronic wounds, implant infections and failure, and multidrug-resistant infections. Aggressive and invasive strategies are employed to cure biofilm infections but are prone to long and expensive treatments, adverse side-effects, and low patient compliance. Recent strategies such as ultrasound-based therapies and antimicrobial nanomaterials have shown some promise in the effective eradication of biofilms. However, maximizing therapeutic effect while minimizing healthy tissue damage is a key challenge that needs to be addressed. Here a combination treatment involving ultrasound and antimicrobial polymeric nanoparticles (PNPs) that synergistically eradicate bacterial biofilms is reported. Ultrasound treatment rapidly disrupts biofilms and increases penetration of antimicrobial PNPs thereby enhancing their antimicrobial activity. This results in superior biofilm toxicity, while allowing for a two- to sixfold reduction in both the concentration of PNPs as well as the duration of ultrasound. Furthermore, that this reduction minimizes cytotoxicity toward fibroblast cells, while resulting in a 100- to 1000-fold reduction in bacterial concentration, is demonstrated.
Collapse
Affiliation(s)
- Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Jessa M V Makabenta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - John J Amante
- Department of Biomedical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA, 01003, USA
| | - Aritra Nath Chattopadhyay
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Dorcas Matuwana
- Department of Biomedical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA, 01003, USA
| | - Cathal J Kearney
- Department of Biomedical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA, 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| |
Collapse
|
44
|
Jara J, Jurado R, Almendro-Vedia VG, López-Montero I, Fernández L, Rodríguez JM, Orgaz B. Interspecies relationships between nosocomial pathogens associated to preterm infants and lactic acid bacteria in dual-species biofilms. Front Cell Infect Microbiol 2022; 12:1038253. [PMID: 36325465 PMCID: PMC9618709 DOI: 10.3389/fcimb.2022.1038253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/26/2022] [Indexed: 12/08/2023] Open
Abstract
The nasogastric enteral feeding tubes (NEFTs) used to feed preterm infants are commonly colonized by bacteria with the ability to form complex biofilms in their inner surfaces. Among them, staphylococci (mainly Staphylococcus epidermidis and Staphylococcus aureus) and some species belonging to the Family Enterobacteriaceae are of special concern since they can cause nosocomial infections in this population. NETF-associated biofilms can also include lactic acid bacteria (LAB), with the ability to compete with pathogenic species for nutrients and space. Ecological interactions among the main colonizers of these devices have not been explored yet; however, such approach could guide future strategies involving the pre-coating of the inner surfaces of NEFTs with well adapted LAB strains in order to reduce the rates of nosocomial infections in neonatal intensive care units (NICUs). In this context, this work implied the formation of dual-species biofilms involving one LAB strain (either Ligilactobacillus salivarius 20SNG2 or Limosilactobacillus reuteri 7SNG3) and one nosocomial strain (either Klebsiella pneumoniae 9SNG3, Serratia marcescens 10SNG3, Staphylococcus aureus 45SNG3 or Staphylococcus epidermidis 46SNG3). The six strains used in this study had been isolated from the inner surface of NEFTs. Changes in adhesion ability of the pathogens were characterized using a culturomic approach. Species interactions and structural changes of the resulting biofilms were analyzed using scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). No aggregation was observed in dual-species biofilms between any of the two LAB strains and either K. pneumoniae 9SNG3 or S. marcescens 10SNG3. In addition, biofilm thickness and volume were reduced, suggesting that both LAB strains can control the capacity to form biofilms of these enterobacteria. In contrast, a positive ecological relationship was observed in the combination L. reuteri 7SNG3-S. aureus 45SNG3. This relationship was accompanied by a stimulation of S. aureus matrix production when compared with its respective monospecies biofilm. The knowledge provided by this study may guide the selection of potentially probiotic strains that share the same niche with nosocomial pathogens, enabling the establishment of a healthier microbial community inside NEFTs.
Collapse
Affiliation(s)
- Josué Jara
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Rubén Jurado
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Víctor G. Almendro-Vedia
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Iván López-Montero
- Departamento de Química Física, Facultad de Químicas, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Madrid, Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
| | - Leonides Fernández
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan Miguel Rodríguez
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Orgaz
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
45
|
Zheng M, Zhu K, Peng H, Shang W, Zhao Y, Lu S, Rao X, Li M, Zhou R, Li G. CcpA Regulates Staphylococcus aureus Biofilm Formation through Direct Repression of Staphylokinase Expression. Antibiotics (Basel) 2022; 11:antibiotics11101426. [PMID: 36290085 PMCID: PMC9598941 DOI: 10.3390/antibiotics11101426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus represents a notorious opportunistic pathogen causing various infections in biofilm nature, imposing remarkable therapeutic challenges worldwide. The catabolite control protein A (CcpA), a major regulator of carbon catabolite repression (CCR), has been recognized to modulate S. aureus biofilm formation, while the underlying mechanism remains to be fully elucidated. In this study, the reduced biofilm was firstly determined in the ccpA deletion mutant of S. aureus clinical isolate XN108 using both crystal violet staining and confocal laser scanning microscopy. RNA-seq analysis suggested that sak-encoding staphylokinase (Sak) was significantly upregulated in the mutant ∆ccpA, which was further confirmed by RT-qPCR. Consistently, the induced Sak production correlated the elevated promoter activity of sak and increased secretion in the supernatants, as demonstrated by Psak-lacZ reporter fusion expression and chromogenic detection, respectively. Notably, electrophoretic mobility shift assays showed that purified recombinant protein CcpA binds directly to the promoter region of sak, suggesting the direct negative control of sak expression by CcpA. Double isogenic deletion of ccpA and sak restored biofilm formation for mutant ∆ccpA, which could be diminished by trans-complemented sak. Furthermore, the exogenous addition of recombinant Sak inhibited biofilm formation for XN108 in a dose-dependent manner. Together, this study delineates a novel model of CcpA-controlled S. aureus biofilm through direct inhibition of sak expression, highlighting the multifaceted roles and multiple networks regulated by CcpA.
Collapse
Affiliation(s)
- Mingxia Zheng
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Keting Zhu
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yan Zhao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Shuguang Lu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Ming Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
- Correspondence: (M.L.); (R.Z.); (G.L.)
| | - Renjie Zhou
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- Correspondence: (M.L.); (R.Z.); (G.L.)
| | - Gang Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
- Correspondence: (M.L.); (R.Z.); (G.L.)
| |
Collapse
|
46
|
Sahoo A, Swain SS, Panda SK, Hussain T, Panda M, Rodrigues CF. In Silico Identification of Potential Insect Peptides against Biofilm-Producing Staphylococcus aureus. Chem Biodivers 2022; 19:e202200494. [PMID: 36198620 DOI: 10.1002/cbdv.202200494] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/13/2022] [Indexed: 11/08/2022]
Abstract
Biofilm-producing Staphylococcus aureus (SA) strains are frequently found in medical environments, from surgical/ wound sites, medical devices. These biofilms reduce the efficacy of applied antibiotics during the treatment of several infections, such as cystic fibrosis, endocarditis, or urinary tract infections. Thus, the development of potential therapeutic agents to destroy the extra protective biofilm layers or to inhibit the biofilm-producing enzymes is urgently needed. Advanced and cost-effective bioinformatics tools are advantageous in locating and speeding up the selection of antibiofilm candidates. Based on the potential drug characteristics, we have selected one-hundred thirty-three antibacterial peptides derived from insects to assess for their antibiofilm potency via molecular docking against five putative biofilm formation and regulated target enzymes: the staphylococcal accessory regulator A or SarA (PDB ID: 2FRH), 4,4'-diapophytoene synthase or CrtM (PDB ID: 2ZCQ), clumping factor A or ClfA (PDB ID: 1N67) and serine-aspartate repeat protein C or SdrC (PDB ID: 6LXH) and sortase A or SrtA (PDB ID: 1T2W) of SA bacterium. In this study, molecular docking was performed using HPEPDOCK and HDOCK servers, and molecular interactions were examined using BIOVIA Discovery Studio Visualizer-2019. The docking score (kcal/mol) range of five promising antibiofilm peptides against five targets was recorded as follows: diptericin A (-215.52 to -303.31), defensin (-201.11 to -301.92), imcroporin (-212.08 to -287.64), mucroporin (-228.72 to -286.76), apidaecin II (-203.90 to -280.20). Among these five, imcroporin and mucroporin were 13 % each, while defensin contained only 1 % of positive net charged residues (Arg+Lys) projected through ProtParam and NetWheels tools. Similarly, imcroporin, mucroporin and apidaecin II were 50 %, while defensin carried 21.05 % of hydrophobic residues predicted by the tool PEPTIDE. 2.0. Most of the peptides exhibited potential characteristics to inhibit S. aureus-biofilm formation via disrupting the cell membrane and cytoplasmic integrity. In summary, the proposed hypothesis can be considered a cost-effective platform for selecting the most promising bioactive drug candidates within a limited timeframe with a greater chance of success in experimental and clinical studies.
Collapse
Affiliation(s)
- Alaka Sahoo
- Department of Skin & VD, Institute of Medical Sciences & SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, 751023 1, India
| | - Shasank S Swain
- Division of Microbiology and NCDs, ICMR-, Regional Medical Research Center, Bhubaneswar, 751023, Odisha, India
| | - Sujogya K Panda
- Center of Environment Climate Change and Public Health, Utkal University, Vani Vihar, Bhubaneswar, 751004, Odisha, India
| | - Tahziba Hussain
- Division of Microbiology and NCDs, ICMR-, Regional Medical Research Center, Bhubaneswar, 751023, Odisha, India
| | - Maitreyee Panda
- Department of Skin & VD, Institute of Medical Sciences & SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, 751023 1, India
| | - Célia F Rodrigues
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário - CESPU, 4585-116 Gandra PRD, Portugal.,LEPABE-Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465, Porto, Portugal.,AliCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| |
Collapse
|
47
|
Ciofu O, Moser C, Jensen PØ, Høiby N. Tolerance and resistance of microbial biofilms. Nat Rev Microbiol 2022; 20:621-635. [PMID: 35115704 DOI: 10.1038/s41579-022-00682-4] [Citation(s) in RCA: 359] [Impact Index Per Article: 179.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
Chronic infections caused by microbial biofilms represent an important clinical challenge. The recalcitrance of microbial biofilms to antimicrobials and to the immune system is a major cause of persistence and clinical recurrence of these infections. In this Review, we present the extent of the clinical problem, and the mechanisms underlying the tolerance of biofilms to antibiotics and to host responses. We also explore the role of biofilms in the development of antimicrobial resistance mechanisms.
Collapse
Affiliation(s)
- Oana Ciofu
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Claus Moser
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Østrup Jensen
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Niels Høiby
- Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
48
|
Antibiofilm Effect of Silver Nanoparticles in Changing the Biofilm-Related Gene Expression of Staphylococcus epidermidis. Int J Mol Sci 2022; 23:ijms23169257. [PMID: 36012520 PMCID: PMC9409202 DOI: 10.3390/ijms23169257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Nowadays, antibiotic resistance is a major public health problem. Among staphylococci, infections caused by Staphylococcus epidermidis (S. epidermidis) are frequent and difficult to eradicate. This is due to its ability to form biofilm. Among the antibiotic substances, nanosilver is of particular interest. Based on this information, we decided to investigate the effect of nanosilver on the viability, biofilm formation and gene expression of the icaADBC operon and the icaR gene for biofilm and non-biofilm S. epidermidis strains. As we observed, the viability of all the tested strains decreased with the use of nanosilver at a concentration of 5 µg/mL. The ability to form biofilm also decreased with the use of nanosilver at a concentration of 3 µg/mL. Genetic expression of the icaADBC operon and the icaR gene varied depending on the ability of the strain to form biofilm. Low concentrations of nanosilver may cause increased biofilm production, however no such effect was observed with high concentrations. This confirms that the use of nanoparticles at an appropriately high dose in any future therapy is of utmost importance. Data from our publication confirm the antibacterial and antibiotic properties of nanosilver. This effect was observed phenotypically and also by levels of gene expression.
Collapse
|
49
|
Wang X, Hong X, Chen F, Wang KJ. A truncated peptide Spgillcin177–189 derived from mud crab Scylla paramamosain exerting multiple antibacterial activities. Front Cell Infect Microbiol 2022; 12:928220. [PMID: 36061863 PMCID: PMC9435603 DOI: 10.3389/fcimb.2022.928220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Antimicrobial peptides (AMPs) may be the most promising substitute for antibiotics due to their effective bactericidal activity and multiple antimicrobial modes against pathogenic bacteria. In this study, a new functional gene named Spgillcin was identified in Scylla paramamosain, which encoded 216 amino acids of mature peptide. In vivo, Spgillcin was dominantly expressed in the gills of male and female crabs, offering the highest expression level among all tested organs or tissues. The expression pattern of Spgillcin was significantly altered when challenged by Staphylococcus aureus, indicating a positive immune response. In vitro, a functional truncated peptide Spgillcin177–189 derived from the amino acid sequence of Spgillcin was synthesized and showed a broad-spectrum and potent antibacterial activity against several bacterial strains, including the clinical isolates of multidrug-resistant (MDR) strains, with a range of minimum inhibitory concentrations from 1.5 to 48 μM. Spgillcin177–189 also showed rapid bactericidal kinetics for S. aureus and Pseudomonas aeruginosa but did not display any cytotoxicity to mammalian cells and maintained its antimicrobial activity in different conditions. Mechanistic studies indicated that Spgillcin177–189 was mainly involved in the disruption of cell membrane integrity where the membrane components lipoteichoic acid and lipopolysaccharide could significantly inhibit the antimicrobial activity in a dose-dependent manner. In addition, Spgillcin177–189 could change the membrane permeability and cause the accumulation of intracellular reactive oxygen species. No resistance was generated to Spgillcin177–189 when the clinical isolates of methicillin-resistant S. aureus and MDR P. aeruginosa were treated with Spgillcin177–189 and then subjected to a long term of continuous culturing for 50 days. In addition, Spgillcin177–189 exerted a strong anti-biofilm activity by inhibiting biofilm formation and was also effective at killing extracellular S. aureus in the cultural supernatant of RAW 264.7 cells. Taken together, Spgillcin177–189 has strong potential as a substitute for antibiotics in future aquaculture and medical applications.
Collapse
Affiliation(s)
- Xiaofei Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Xiao Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- *Correspondence: Ke-Jian Wang,
| |
Collapse
|
50
|
Sionov RV, Banerjee S, Bogomolov S, Smoum R, Mechoulam R, Steinberg D. Targeting the Achilles' Heel of Multidrug-Resistant Staphylococcus aureus by the Endocannabinoid Anandamide. Int J Mol Sci 2022; 23:7798. [PMID: 35887146 PMCID: PMC9319909 DOI: 10.3390/ijms23147798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Antibiotic-resistant Staphylococcus aureus is a major health issue that requires new therapeutic approaches. Accumulating data suggest that it is possible to sensitize these bacteria to antibiotics by combining them with inhibitors targeting efflux pumps, the low-affinity penicillin-binding protein PBP2a, cell wall teichoic acid, or the cell division protein FtsZ. We have previously shown that the endocannabinoid Anandamide (N-arachidonoylethanolamine; AEA) could sensitize drug-resistant S. aureus to a variety of antibiotics, among others, through growth arrest and inhibition of drug efflux. Here, we looked at biochemical alterations caused by AEA. We observed that AEA increased the intracellular drug concentration of a fluorescent penicillin and augmented its binding to membrane proteins with concomitant altered membrane distribution of these proteins. AEA also prevented the secretion of exopolysaccharides (EPS) and reduced the cell wall teichoic acid content, both processes known to require transporter proteins. Notably, AEA was found to inhibit membrane ATPase activity that is necessary for transmembrane transport. AEA did not affect the membrane GTPase activity, and the GTPase cell division protein FtsZ formed the Z-ring of the divisome normally in the presence of AEA. Rather, AEA caused a reduction in murein hydrolase activities involved in daughter cell separation. Altogether, this study shows that AEA affects several biochemical processes that culminate in the sensitization of the drug-resistant bacteria to antibiotics.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Shreya Banerjee
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Sergei Bogomolov
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Reem Smoum
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (R.S.); (R.M.)
| | - Raphael Mechoulam
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (R.S.); (R.M.)
| | - Doron Steinberg
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| |
Collapse
|