101
|
Sharma DK, Rajpurohit YS. Multitasking functions of bacterial extracellular DNA in biofilms. J Bacteriol 2024; 206:e0000624. [PMID: 38445859 PMCID: PMC11025335 DOI: 10.1128/jb.00006-24] [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] [Indexed: 03/07/2024] Open
Abstract
Bacterial biofilms are intricate ecosystems of microbial communities that adhere to various surfaces and are enveloped by an extracellular matrix composed of polymeric substances. Within the context of bacterial biofilms, extracellular DNA (eDNA) originates from cell lysis or is actively secreted, where it exerts a significant influence on the formation, stability, and resistance of biofilms to environmental stressors. The exploration of eDNA within bacterial biofilms holds paramount importance in research, with far-reaching implications for both human health and the environment. An enhanced understanding of the functions of eDNA in biofilm formation and antibiotic resistance could inspire the development of strategies to combat biofilm-related infections and improve the management of antibiotic resistance. This comprehensive review encapsulates the latest discoveries concerning eDNA, encompassing its origins, functions within bacterial biofilms, and significance in bacterial pathogenesis.
Collapse
Affiliation(s)
- Dhirendra Kumar Sharma
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India
- Schools of Life Sciences, Homi Bhabha National Institute (DAE—Deemed University), Mumbai, India
| | - Yogendra Singh Rajpurohit
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India
- Schools of Life Sciences, Homi Bhabha National Institute (DAE—Deemed University), Mumbai, India
| |
Collapse
|
102
|
Araújo D, Silva AR, Fernandes R, Serra P, Barros MM, Campos AM, Oliveira R, Silva S, Almeida C, Castro J. Emerging Approaches for Mitigating Biofilm-Formation-Associated Infections in Farm, Wild, and Companion Animals. Pathogens 2024; 13:320. [PMID: 38668275 PMCID: PMC11054384 DOI: 10.3390/pathogens13040320] [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: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
The importance of addressing the problem of biofilms in farm, wild, and companion animals lies in their pervasive impact on animal health and welfare. Biofilms, as resilient communities of microorganisms, pose a persistent challenge in causing infections and complicating treatment strategies. Recognizing and understanding the importance of mitigating biofilm formation is critical to ensuring the welfare of animals in a variety of settings, from farms to the wild and companion animals. Effectively addressing this issue not only improves the overall health of individual animals, but also contributes to the broader goals of sustainable agriculture, wildlife conservation, and responsible pet ownership. This review examines the current understanding of biofilm formation in animal diseases and elucidates the complex processes involved. Recognizing the limitations of traditional antibiotic treatments, mechanisms of resistance associated with biofilms are explored. The focus is on alternative therapeutic strategies to control biofilm, with illuminating case studies providing valuable context and practical insights. In conclusion, the review highlights the importance of exploring emerging approaches to mitigate biofilm formation in animals. It consolidates existing knowledge, highlights gaps in understanding, and encourages further research to address this critical facet of animal health. The comprehensive perspective provided by this review serves as a foundation for future investigations and interventions to improve the management of biofilm-associated infections in diverse animal populations.
Collapse
Affiliation(s)
- Daniela Araújo
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Ana Rita Silva
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Rúben Fernandes
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Patrícia Serra
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Maria Margarida Barros
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CECAV—Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Ana Maria Campos
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Ricardo Oliveira
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sónia Silva
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Carina Almeida
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana Castro
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
103
|
Bandeira DM, Corrêa JM, Laskoski LV, Rosset J, Conceição LHSM, Gomes SD, Pinto FGS. Phytochemical screening of Podocarpus lambertii Klotzch ex Endl. leaf extracts and potential antimicrobial, antioxidant and antibiofilm activity. AN ACAD BRAS CIENC 2024; 96:e20230237. [PMID: 38655919 DOI: 10.1590/0001-3765202420230237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/26/2023] [Indexed: 04/26/2024] Open
Abstract
Species of the genus Podocarpus L'Hér. ex Pers.present biological activities, such as analgesic, antioxidant, antifungal, acting in the fight against anemia, depurative and fortifying. Podocarpus lambertii Klotzch ex Endl. is a Brazilian native species popularly known as maritime pine and lacks information about its phytochemical profile and possible biological activities. The study was conducted to determine the phytochemical composition of soluble plant extracts of acetone (EA), ethyl acetate (EAE) and hexane (HE) from leaves of P. lambertii; evaluate the antimicrobial potential by the broth microdilution technique; antioxidant potential by the DPPH method, as well as to evaluate the biofilm inhibition capacity by the crystal violet assay and reduction of the yellow tetrazolium salt (MTT). Phytochemical screening detected the presence of flavonoids, triterpenoids, steroids, tannins, alkaloids and saponins. All extracts showed antimicrobial activity on the microorganisms tested, and the EA showed the best results. High free radical scavenging potential was observed only in EAE (96.35%). The antibiofilm potential was observed in the EAE extract. The results contribute to the knowledge of the species and indicate the potential of P. lambertii extracts as a source of plant bioactives for the development of new alternative strategies to control resistant microorganisms.
Collapse
Affiliation(s)
- Debora Marina Bandeira
- Programa de Pós-Graduação Stricto Sensu em Conservação e Manejo de Recursos Naturais, Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia -LAMIBI, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
- Programa de Pós-Graduação em Engenharia Agrícola, Universidade Estadual do Oeste do Paraná, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| | - Juliana M Corrêa
- Programa de Pós-Graduação Stricto Sensu em Conservação e Manejo de Recursos Naturais, Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia -LAMIBI, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| | - Larissa Valéria Laskoski
- Programa de Pós-Graduação Stricto Sensu em Conservação e Manejo de Recursos Naturais, Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia -LAMIBI, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| | - Jéssica Rosset
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| | - Lázaro Henrique S M Conceição
- Programa de Pós-Graduação Stricto Sensu em Conservação e Manejo de Recursos Naturais, Universidade Estadual do Oeste do Paraná, Herbário HUOP, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| | - Simone D Gomes
- Programa de Pós-Graduação em Engenharia Agrícola, Universidade Estadual do Oeste do Paraná, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| | - Fabiana Gisele S Pinto
- Programa de Pós-Graduação Stricto Sensu em Conservação e Manejo de Recursos Naturais, Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia -LAMIBI, Rua Universitária, 2069, 85819-110 Cascavel, PR, Brazil
| |
Collapse
|
104
|
Ekhtiari-Sadegh S, Samani S, Barneh F, Dashtbin S, Shokrgozar MA, Pooshang Bagheri K. Rapid eradication of vancomycin and methicillin-resistant Staphylococcus aureus by MDP1 antimicrobial peptide coated on photocrosslinkable chitosan hydrogel: in vitro antibacterial and in silico molecular docking studies. Front Bioeng Biotechnol 2024; 12:1385001. [PMID: 38681961 PMCID: PMC11047131 DOI: 10.3389/fbioe.2024.1385001] [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: 02/11/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction Antibiotic resistance and weak bioavailability of antibiotics in the skin due to systemic administration leads to failure in eradication of vancomycin- and methicillin-resistant Staphylococcus aureus (VRSA and MRSA)-associated wound infections and subsequent septicemia and even death. Accordingly, this study aimed at designing a photocrosslinkable methacrylated chitosan (MECs) hydrogel coated by melittin-derived peptide 1 (MDP1) that integrated the antibacterial activity with the promising skin regenerative capacity of the hydrogel to eradicate bacteria by burst release strategy. Methods The MECs was coated with MDP1 (MECs-MDP1), characterized, and the hydrogel-peptide interaction was evaluated by molecular docking. Antibacterial activities of MECs-MDP1 were evaluated against VRSA and MRSA bacteria and compared to MECs-vancomycin (MECs-vanco). Antibiofilm activity of MECs-MDP1 was studied by our novel 'in situ biofilm inhibition zone (IBIZ)' assay, and SEM. Biocompatibility with human dermal fibroblast cells (HDFs) was also evaluated. Results and Discussion Molecular docking showed hydrogen bonds as the most interactions between MDP1 and MECs at a reasonable affinity. MECs-MDP1 eradicated the bacteria rapidly by burst release strategy whereas MECs-vanco failed to eradicate them at the same time intervals. Antibiofilm activity of MECs-MDP1 were also proved successfully. As a novel report, molecular docking analysis has demonstrated that MDP1 covers the structure of MECs and also binds to lysozyme with a reasonable affinity, which may explain the inhibition of lysozyme. MECs-MDP1 was also biocompatible with human dermal fibroblast skin cells, which indicates its safe future application. The antibacterial properties of a photocrosslinkable methacrylated chitosan-based hydrogel coated with MDP1 antimicrobial peptide were successfully proved against the most challenging antibiotic-resistant bacteria causing nosocomial wound infections; VRSA and MRSA. Molecular docking analysis revealed that MDP1 interacts with MECs mainly through hydrogen bonds with reasonable binding affinity. MECs-MDP1 hydrogels eradicated the planktonic state of bacteria by burst release of MDP1 in just a few hours whereas MECs-vanco failed to eradicate them. inhibition zone assay showed the anti-biofilm activity of the MECs-MDP1 hydrogel too. These findings emphasize that MECs-MDP1 hydrogel would be suggested as a biocompatible wound-dressing candidate with considerable and rapid antibacterial activities to prevent/eradicate VRSA/MRSA bacterial wound infections.
Collapse
Affiliation(s)
- Sarvenaz Ekhtiari-Sadegh
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Saeed Samani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnoosh Barneh
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shirin Dashtbin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
105
|
Blair N, Patil P, Nguyen D, Paudyal-Nepal B, Iorember F. Antibiotic lock solutions as adjunct therapy for catheter-related blood stream infections in pediatric hemodialysis patients. Front Pediatr 2024; 12:1379895. [PMID: 38665376 PMCID: PMC11043483 DOI: 10.3389/fped.2024.1379895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The predominant use of intravenous catheters as primary access type in the pediatric hemodialysis population is associated with an increased risk of catheter related blood stream infections. While strict adherence to catheter placement and long-term care guidelines have helped to decrease the incidence of these infections, blood stream infections remain an infection burden in pediatric patients with long term hemodialysis catheters. The formation of biofilms on the surfaces of these catheters has been shown to be a source of microbes causing blood stream infections. One of the strategies for preventing bacterial colonization, inhibiting microbial multiplication, and suppressing the seeding of these microbes from biofilms upon maturation, has been the use of antibiotic-based lock solutions in-between dialysis treatments. Although clinical guidelines for the use of antibiotic lock solutions are yet to be developed, available evidence suggests a beneficial role of antibiotic lock solutions in the management of catheter related blood stream infections. Additionally, a clear understanding of how biofilms are formed and their role in the pathogenesis of catheter related bloodstream infection will facilitate the development of solutions that can prevent biofilm formation and inhibit their multiplication, maturation and seeding into the bloodstream.
Collapse
Affiliation(s)
- N. Blair
- School of Medicine, Texas A&M University School of Medicine, College Station, TX, United States
| | - P. Patil
- Department of Pharmacy, Driscoll Children’s Hospital, Corpus Christi, TX, United States
| | - D. Nguyen
- Department of Pharmacy, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - B. Paudyal-Nepal
- Department of Nephrology, Texas Children’s Hospital, Austin, TX, United States
| | - F. Iorember
- Department of Nephrology, Driscoll Children's Hospital, Corpus Christi, TX, United States
| |
Collapse
|
106
|
Aleksandrova YI, Shurpik DN, Nazmutdinova VA, Zelenikhin PV, Subakaeva EV, Sokolova EA, Leonteva YO, Mironova AV, Kayumov AR, Petrovskii VS, Potemkin II, Stoikov II. Antibacterial Activity of Various Morphologies of Films Based on Guanidine Derivatives of Pillar[5]arene: Influence of the Nature of One Substitute on Self-assembly. ACS APPLIED MATERIALS & INTERFACES 2024; 16:17163-17181. [PMID: 38530408 DOI: 10.1021/acsami.3c18610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
The progress of the pillar[5]arene chemistry allowed us to set out a new concept on application of the supramolecular assemblies to create antimicrobial films with variable surface morphologies and biological activities. Antibacterial films were derived from the substituted pillar[5]arenes containing nine pharmacophoric guanidine fragments and one thioalkyl substituent. Changing the only thioalkyl fragment in the macrocycle structure made it possible to control the biological activity of the resulting antibacterial coating. Pretreatment of the surface with aqueous solution of the amphiphilic pillar[5]arenes reduced the biofilm thickness by 56 ± 10% of Gram-positive Staphylococcus aureus in the case of the pillar[5]arene containing a thiooctyl fragment and by 52 ± 7% for the biofilm of Gram-negative Klebsiella pneumoniae in the case of pillar[5]arene containing a thiooctadecyl fragment. Meanwhile, the cytotoxicity of the synthesized macrocycles was examined at a concentration of 50 μg/mL, which was significantly lower than that of bis-guanidine-based antimicrobial preparations.
Collapse
Affiliation(s)
- Yulia I Aleksandrova
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 420008 Kremlevskaya Street, 18, Kazan, Russian Federation
| | - Dmitriy N Shurpik
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 420008 Kremlevskaya Street, 18, Kazan, Russian Federation
| | - Viktoriya A Nazmutdinova
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 420008 Kremlevskaya Street, 18, Kazan, Russian Federation
| | - Pavel V Zelenikhin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kremlevskaya, 18, Kazan, Russian Federation
| | - Evgeniya V Subakaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kremlevskaya, 18, Kazan, Russian Federation
| | - Evgeniya A Sokolova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kremlevskaya, 18, Kazan, Russian Federation
| | - Yulia O Leonteva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kremlevskaya, 18, Kazan, Russian Federation
| | - Anna V Mironova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kremlevskaya, 18, Kazan, Russian Federation
| | - Airat R Kayumov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kremlevskaya, 18, Kazan, Russian Federation
| | - Vladislav S Petrovskii
- Physics Department, M. V. Lomonosov Moscow State University, Leninskie Gory 1-2, 119991 Moscow, Russian Federation
- N. N. Semenov Federal Research Center of Chemical Physics of Russian Academy of Sciences, Kosygina 4, 119991 Moscow, Russian Federation
| | - Igor I Potemkin
- Physics Department, M. V. Lomonosov Moscow State University, Leninskie Gory 1-2, 119991 Moscow, Russian Federation
| | - Ivan I Stoikov
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 420008 Kremlevskaya Street, 18, Kazan, Russian Federation
| |
Collapse
|
107
|
Wang L, Zheng J, Hou W, Zhang C, Zhang J, Fan X, Zhang H, Han Y. The Anti-Microbial Peptide Citrocin Controls Pseudomonas aeruginosa Biofilms by Breaking Down Extracellular Polysaccharide. Int J Mol Sci 2024; 25:4122. [PMID: 38612931 PMCID: PMC11012989 DOI: 10.3390/ijms25074122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Citrocin is an anti-microbial peptide that holds great potential in animal feed. This study evaluates the anti-microbial and anti-biofilm properties of Citrocin and explores the mechanism of action of Citrocin on the biofilm of P. aeruginosa. The results showed that Citrocin had a significant inhibitory effect on the growth of P. aeruginosa with a minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of 0.3 mg/mL. All five concentrations (1/4MIC, 1/2MIC, MIC, 2MIC, and 4MIC) of Citrocin inhibited P. aeruginosa biofilm formation. Citrocin at the MIC, 2MIC and 4MIC removed 42.7%, 76.0% and 83.2% of mature biofilms, respectively, and suppressed the swarming motility, biofilm metabolic activity and extracellular polysaccharide production of P. aeruginosa. Metabolomics analysis indicated that 0.3 mg/mL of Citrocin up- regulated 26 and down-regulated 83 metabolites, mainly comprising amino acids, fatty acids, organic acids and sugars. Glucose and amino acid metabolic pathways, including starch and sucrose metabolism as well as arginine and proline metabolism, were highly enriched by Citrocin. In summary, our research reveals the anti-biofilm mechanism of Citrocin at the metabolic level, which provides theoretical support for the development of novel anti-biofilm strategies for combatting P. aeruginosa.
Collapse
Affiliation(s)
- Liyao Wang
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
- College of Life Science and Technology, Southeast University, Nanjing 211189, China
| | - Jiaqi Zheng
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wenchao Hou
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
| | - Chaowen Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
| | - Jie Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
| | - Xuanbo Fan
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
| | - Hongliang Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
- College of Animal Science and Technology, China Agricultural University, Beijing 100083, China
| | - Yuzhu Han
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China; (L.W.); (J.Z.); (W.H.); (C.Z.); (J.Z.); (X.F.); (H.Z.)
- Chongqing Key Laboratory of Herbivore Science, Chongqing 402460, China
| |
Collapse
|
108
|
Kunz Coyne AJ, Stamper K, Bleick C, Kebriaei R, Lehman SM, Rybak MJ. Synergistic bactericidal effects of phage-enhanced antibiotic therapy against MRSA biofilms. Microbiol Spectr 2024; 12:e0321223. [PMID: 38411110 PMCID: PMC10986480 DOI: 10.1128/spectrum.03212-23] [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] [Received: 08/30/2023] [Accepted: 01/10/2024] [Indexed: 02/28/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes biofilm-related medical device infections. Phage-antibiotic combinations offer potential therapy due to proven in vitro antibiofilm efficacy. We evaluated phage-antibiotic synergy against biofilms using modified checkerboard and 24-h time-kill assays. Humanized-simulated daptomycin (DAP) (10, 8, and 6 mg/kg q24h) and ceftaroline (CPT) (600 mg q12h) were combined with Intesti13, Sb-1, and Romulus phages (tMOI 1, q12h). Assays were conducted in 168-h biofilm reactor models against DAP non-susceptible (DNS) vancomycin intermediate S. aureus (VISA) MRSA D712 and DAP-susceptible MRSA 8014. Synergistic activity and bactericidal activity were defined as ≥2log10 CFU/mL reduction from antibiotic-only regimens and ≥3log10 CFU/mL decrease from baseline at 24 h. Differences were analyzed by one-way analysis of variance with Tukey's post hoc test (P ≤ 0.05 is considered significant). Surviving bacteria were examined for antibiotic minimum biofilm inhibitory concentration (MBIC) changes and phage susceptibility. In 168-h biofilm models, humanized DAP 10 mg/kg + CPT, combined with a 2-phage cocktail (Intesti13 + Sb-1) against D712, and a 3-phage cocktail (Intesti13 + Sb-1 + Romulus) against 8014, demonstrated synergistic bactericidal activity. At 168 h, bacteria were minimally detectable [2log10 CFU/cm2 (-Δ4.23 and -Δ4.42 log10 CFU/cm2; both P < 0.001)]. Antibiotic MBIC remained unchanged compared to baseline across various time points. None of the tested bacteria at 168 h exhibited complete phage resistance. This study reveals bactericidal efficacy of DAP + CPT with 2-phage and 3-phage cocktails against DNS VISA and MRSA isolates (D712 and 8014) in biofilm models, maintaining susceptibility. Further research is needed for diverse strains and durations, aligning with infection care. IMPORTANCE The prevalence of biofilm-associated medical device infections caused by methicillin-resistant Staphylococcus aureus (MRSA) presents a pressing medical challenge. The latest research demonstrates the potential of phage-antibiotic combinations (PACs) as a promising solution, notably in vitro antibiofilm efficacy. By adopting modified checkerboard and 24-h time-kill assays, the study investigated the synergistic action of phages combined with humanized-simulated doses of daptomycin (DAP) and ceftaroline (CPT). The results were promising: a combination of DAP, CPT, and either a 2-phage or 3-phage cocktail effectively exhibited bactericidal activity against both DAP non-susceptible vancomycin intermediate S. aureus MRSA and DAP-susceptible MRSA strains within 168-h biofilm models. Moreover, post-treatment evaluations revealed no discernible rise in antibiotic resistance or complete phage resistance. This pioneering work suggests the potential of PACs in addressing MRSA biofilm infections, setting the stage for further expansive research tailored to diverse bacterial strains and treatment durations.
Collapse
Affiliation(s)
- Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kyle Stamper
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Callan Bleick
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Razieh Kebriaei
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Susan M. Lehman
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- Department of Pharmacy Services, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA
- Department of Medicine, Division of Infectious Diseases, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
109
|
Fang ZY, Zhang ZY, Zheng YD, Lei D, Zhuang J, Li N, He QY, Sun X. Repurposing cinacalcet suppresses multidrug-resistant Staphylococcus aureus by disruption of cell membrane and inhibits biofilm by targeting IcaR. J Antimicrob Chemother 2024; 79:903-917. [PMID: 38412335 DOI: 10.1093/jac/dkae051] [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] [Received: 09/01/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND MDR Staphylococcus aureus infections, along with the severity of biofilm-associated infections, continue to threaten human health to a great extent. It necessitates the urgent development of novel antimicrobial and antibiofilm agents. OBJECTIVES To reveal the mechanism and target of cinacalcet as an antibacterial and antimicrobial agent for S. aureus. METHODS Screening of non-antibiotic drugs for antibacterial and antibiofilm properties was conducted using a small-molecule drug library. In vivo efficacy was assessed through animal models, and the antibacterial mechanism was studied using quantitative proteomics, biochemical assays, LiP-SMap, BLI detection and gene knockout techniques. RESULTS Cinacalcet, an FDA-approved drug, demonstrated antibacterial and antibiofilm activity against S. aureus, with less observed development of bacterial resistance. Importantly, cinacalcet significantly improved survival in a pneumonia model and bacterial clearance in a biofilm infection model. Moreover, the antibacterial mechanism of cinacalcet mainly involves the destruction of membrane-targeted structures, alteration of energy metabolism, and production of reactive oxygen species (ROS). Cinacalcet was found to target IcaR, inhibiting biofilm formation through the negative regulation of IcaADBC. CONCLUSIONS The findings suggest that cinacalcet has potential for repurposing as a therapeutic agent for MDR S. aureus infections and associated biofilms, warranting further investigation.
Collapse
Affiliation(s)
- Zu-Ye Fang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zi-Yuan Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yun-Dan Zheng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dan Lei
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jianpeng Zhuang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Nan Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xuesong Sun
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
110
|
Hofer LK, Jurcisek JA, Elmaraghy C, Goodman SD, Bakaletz LO. Z-Form Extracellular DNA in Pediatric CRS May Provide a Mechanism for Recalcitrance to Treatment. Laryngoscope 2024; 134:1564-1571. [PMID: 37597166 PMCID: PMC10875147 DOI: 10.1002/lary.30986] [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: 05/19/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
OBJECTIVES We examined sinus mucosal samples recovered from pediatric chronic rhinosinusitis (CRS) patients for the presence of Z-form extracellular DNA (eDNA) due to its recently elucidated role in pathogenesis of disease. Further, we immunolabeled these specimens for the presence of both members of the bacterial DNA-binding DNABII protein family, integration host factor (IHF) and histone-like protein (HU), due to their known role in converting common B-DNA to the rare Z-form. METHODS Sinus mucosa samples recovered from 20 patients during functional endoscopic sinus surgery (FESS) were immunolabelled for B- and Z-DNA, as well as for both bacterial DNABII proteins. RESULTS Nineteen of 20 samples (95%) included areas rich in eDNA, with the majority in the Z-form. Areas positive for B-DNA were restricted to the most distal regions of the mucosal specimen. Labeling for both DNABII proteins was observed on B- and Z-DNA, which aligned with the role of these proteins in the B-to-Z DNA conversion. CONCLUSIONS Abundant Z-form eDNA in culture-positive pediatric CRS samples suggested that bacterial DNABII proteins were responsible for the conversion of eukaryotic B-DNA that had been released into the luminal space by PMNs during NETosis, to the Z-form. The presence of both DNABII proteins on B-DNA and Z-DNA supported the known role of these bacterial proteins in the B-to-Z DNA conversion. Given that Z-form DNA both stabilizes the bacterial biofilm and inactivates PMN NET-mediated killing of trapped bacteria, we hypothesize that this conversion may be contributing to the chronicity and recalcitrance of CRS to treatment. LEVEL OF EVIDENCE NA Laryngoscope, 134:1564-1571, 2024.
Collapse
Affiliation(s)
- Llwyatt K. Hofer
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Joseph A. Jurcisek
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Charles Elmaraghy
- The Ohio State University College of Medicine
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Wexner Medical Center
- Department of Pediatric Otolaryngology, Nationwide Children’s Hospital
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital
- The Ohio State University College of Medicine
| | - Lauren O. Bakaletz
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital
- The Ohio State University College of Medicine
| |
Collapse
|
111
|
Lou Y, Palermo EF. Dynamic Antimicrobial Poly(disulfide) Coatings Exfoliate Biofilms On Demand Via Triggered Depolymerization. Adv Healthc Mater 2024; 13:e2303359. [PMID: 38288658 DOI: 10.1002/adhm.202303359] [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: 11/08/2023] [Indexed: 02/13/2024]
Abstract
Bacterial biofilms are notoriously problematic in applications ranging from biomedical implants to ship hulls. Cationic, amphiphilic antibacterial surface coatings delay the onset of biofilm formation by killing microbes on contact, but they lose effectiveness over time due to non-specific binding of biomass and biofilm formation. Harsh treatment methods are required to forcibly expel the biomass and regenerate a clean surface. Here, a simple, dynamically reversible method of polymer surface coating that enables both chemical killing on contact, and on-demand mechanical delamination of surface-bound biofilms, by triggered depolymerization of the underlying antimicrobial coating layer, is developed. Antimicrobial polymer derivatives based on α-lipoic acid (LA) undergo dynamic and reversible polymerization into polydisulfides functionalized with biocidal quaternary ammonium salt groups. These coatings kill >99.9% of Staphylococcus aureus cells, repeatedly for 15 cycles without loss of activity, for moderate microbial challenges (≈105 colony-forming units (CFU) mL-1, 1 h), but they ultimately foul under intense challenges (≈107 CFU mL-1, 5 days). The attached biofilms are then exfoliated from the polymer surface by UV-triggered degradation in an aqueous solution at neutral pH. This work provides a simple strategy for antimicrobial coatings that can kill bacteria on contact for extended timescales, followed by triggered biofilm removal under mild conditions.
Collapse
Affiliation(s)
- Yang Lou
- Materials Science and Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY, 12180, USA
| | - Edmund F Palermo
- Materials Science and Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY, 12180, USA
- Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY, 12180, USA
| |
Collapse
|
112
|
Pu F, Fang J, Li W, Zhang B, Hong X, Xu L, Li X, Jiang Y. New Alpiniamide-Type Polyketide with Antibiofilm Activities from the Marine-Derived Streptomyces sp. ZS-A65. Chem Biodivers 2024; 21:e202400029. [PMID: 38270294 DOI: 10.1002/cbdv.202400029] [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: 01/05/2024] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 01/26/2024]
Abstract
Two new alpiniamide-type polyketides, alpiniamides H-I (1-2), in addition to four recognized compounds, were discovered in Streptomyces sp. ZSA65 derived from the marine sediments. The planar structure and absolute configuration of alpiniamides H-I were elucidated using a combination of 1D, 2D NMR, HRESIMS data analysis, Mosher's method and ECD calculations. The antibiofilm and antibacterial activities against P. aeruginosa were evaluated using the microdilution method. Notably, Compound 2 exhibited strong antibiofilm property.
Collapse
Affiliation(s)
- Fanqi Pu
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| | - Jiebin Fang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, People's Republic of China
| | - Wenshuo Li
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| | - Bin Zhang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| | - Xinyi Hong
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| | - Lei Xu
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| | - Xihui Li
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| | - Yongjun Jiang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316021, People's Republic of China
| |
Collapse
|
113
|
Blee JA, Gorochowski TE, Hauert S. Optimization of periodic treatment strategies for bacterial biofilms using an agent-based in silico approach. J R Soc Interface 2024; 21:20240078. [PMID: 38593842 PMCID: PMC11003776 DOI: 10.1098/rsif.2024.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Biofilms are responsible for most chronic infections and are highly resistant to antibiotic treatments. Previous studies have demonstrated that periodic dosing of antibiotics can help sensitize persistent subpopulations and reduce the overall dosage required for treatment. Because the dynamics and mechanisms of biofilm growth and the formation of persister cells are diverse and are affected by environmental conditions, it remains a challenge to design optimal periodic dosing regimens. Here, we develop a computational agent-based model to streamline this process and determine key parameters for effective treatment. We used our model to test a broad range of persistence switching dynamics and found that if periodic antibiotic dosing was tuned to biofilm dynamics, the dose required for effective treatment could be reduced by nearly 77%. The biofilm architecture and its response to antibiotics were found to depend on the dynamics of persister cells. Despite some differences in the response of biofilm governed by different persister switching rates, we found that a general optimized periodic treatment was still effective in significantly reducing the required antibiotic dose. As persistence becomes better quantified and understood, our model has the potential to act as a foundation for more effective strategies to target bacterial infections.
Collapse
Affiliation(s)
- Johanna A. Blee
- School of Engineering Mathematics and Technology, University of Bristol, Ada Lovelace Building, Tankard's Close, Bristol BS8 1TW, UK
- School of Biological Sciences, University of Bristol, Life Sciences Building, 24 Tyndall Avenue, Bristol BS8 1TQ, UK
| | - Thomas E. Gorochowski
- School of Biological Sciences, University of Bristol, Life Sciences Building, 24 Tyndall Avenue, Bristol BS8 1TQ, UK
- BrisEngBio, School of Chemistry, University of Bristol, Cantock's Close, Bristol BS8 1TS, UK
| | - Sabine Hauert
- School of Engineering Mathematics and Technology, University of Bristol, Ada Lovelace Building, Tankard's Close, Bristol BS8 1TW, UK
- BrisEngBio, School of Chemistry, University of Bristol, Cantock's Close, Bristol BS8 1TS, UK
| |
Collapse
|
114
|
Yun S, Huang J, Zhang M, Wang X, Wang X, Zhou Y. Preliminary identification and semi-quantitative characterization of a multi-faceted high-stability alginate lyase from marine microbe Seonamhaeicola algicola with anti-biofilm effect on Pseudomonas aeruginosa. Enzyme Microb Technol 2024; 175:110408. [PMID: 38309052 DOI: 10.1016/j.enzmictec.2024.110408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Alginate lyases with unique characteristics for degrading alginate into size-defined oligosaccharide fractions, were considered as the potential agents for disrupting Pseudomonas aeruginosa biofilms. In our study, a novel endolytic PL-7 alginate lyase, named AlyG2, was cloned and expressed through Escherichia coli. This enzyme exhibited excellent properties: it maintained more than 85% activity at low temperatures of 4 °C and high temperatures of 70 °C. After 1 h of incubation at 4 °C, it still retained over 95% activity, demonstrating the ability to withstand low temperature. The acid-base and salt tolerance properties shown it preserves more than 50% activity in the pH range of 5.0 to 11.0 and in a high salt environment at 3000 mM NacCl, indicating its high stability in several aspects. More importantly, AlyG2 in our research was revealed to be effective at removing mature biofilms and inhibiting biofilm formation produced by Pseudomonas aeruginosa, and the inhibition and disruption rates were 47.25 ± 4.52% and 26.5 ± 6.72%, respectively. Additionally, the enzyme AlyG2 promoted biofilm disruption in combination with antibiotics, particularly manifesting the synergistic effect with erythromycin (FIC=0.5). In all, these results offered that AlyG2 with unique characteristics may be an effective technique for the clearance or disruption of biofilm produced by P. aeruginosa.
Collapse
Affiliation(s)
- Shuaiting Yun
- Marine College, Shandong University, Weihai 264209, China
| | - Jinping Huang
- Marine College, Shandong University, Weihai 264209, China
| | - Mingjing Zhang
- Marine College, Shandong University, Weihai 264209, China
| | - Xueting Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Xiaochen Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Yanxia Zhou
- Marine College, Shandong University, Weihai 264209, China.
| |
Collapse
|
115
|
Laskoski LV, Batista JM, Bandeira DM, Corrêa JM, Rosset J, Conceição LHSM, Pinto FGS. Investigation of phytochemical composition and bioactivity evaluation of extracts from Myrsine umbellata Mart. BRAZ J BIOL 2024; 84:e276871. [PMID: 38451630 DOI: 10.1590/1519-6984.276871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 02/12/2024] [Indexed: 03/08/2024] Open
Abstract
The objective of the study was to carry out phytochemical prospection through colorimetric tests to determine the groups of secondary metabolites and also to determine the total content of phenolic compounds (TPC) present in plant extracts methanol (ME), ethyl acetate (EAE), hexane (HE) and dichloromethane (DE) from the leaves of Myrsine umbellata, as well as to investigate the antimicrobial activity against twelve standard ATCC strains by the broth microdilution technique; the antioxidant potential by the DPPH method and the ABTS method and the antibiofilm potential on the biofilm biomass of standard bacteria by the crystal violet technique and tetrazolium salt reduction (MTT) assay. Phytochemical prospection detected the presence of saponins, steroids, alkaloids, anthocyanins, anthocyanidins, flavonoids, and tannins. The results of the quantitative phytochemical estimation revealed a higher content of total phenolics in DE (280.24 ± 0.037 µM GAE g ext. -1) followed by ME (159.01 ± 0.031 µM GAE g ext. -1). The ME showed the best biological activities when compared to the other extracts tested. We observed antimicrobial activity against Gram-positive Staphylococcus epidermidis strain (MIC 3.12 and MBC 6.25), antioxidant percentage of 92.58% against the DPPH radical and 420.31 µM Trolox g ext. -1 against the ABTS radical, finally showed antibiofilm action against Gram-positive strain Staphylococcus aureus, with eradication of the biomass in 92.58%. The results suggest that EM from M. umbellata represents an alternative source of plant bioactives for the development of natura products.
Collapse
Affiliation(s)
- L V Laskoski
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia - LAMIBI, Cascavel, PR, Brasil
| | - J M Batista
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia - LAMIBI, Cascavel, PR, Brasil
| | - D M Bandeira
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia - LAMIBI, Cascavel, PR, Brasil
| | - J M Corrêa
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia - LAMIBI, Cascavel, PR, Brasil
| | - J Rosset
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia - LAMIBI, Cascavel, PR, Brasil
| | - L H S M Conceição
- Universidade Estadual do Oeste do Paraná, UNOP Herbário, Cascavel, PR, Brasil
| | - F G S Pinto
- Universidade Estadual do Oeste do Paraná, Laboratório de Microbiologia e Biotecnologia - LAMIBI, Cascavel, PR, Brasil
| |
Collapse
|
116
|
Elumalai R, Vishwakarma A, Balakrishnan A, Ramya M. Assessment of the growth inhibition and anti-biofilm activity of aptamer (PmA2G02) against Proteus mirabilis 1429 T. Res Microbiol 2024; 175:104105. [PMID: 37429429 DOI: 10.1016/j.resmic.2023.104105] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/15/2023] [Accepted: 07/01/2023] [Indexed: 07/12/2023]
Abstract
Proteus mirabilis is known to cause Catheter-associated urinary tract infections (CAUTIs), which exhibit virulence factors linked to forming biofilms. Aptamers have recently been explored as potential anti-biofilm agents. This study demonstrates the anti-biofilm activity of aptamer (PmA2G02) targeting P. mirabilis 1429T, a pathogenic bacteria known to cause Catheter-associated urinary tract infections (CAUTIs). The studied aptamer inhibited biofilm formation, swarming motility, and cell viability at a concentration of 3 μM. The study also showed that the PmA2G02 had a binding affinity towards fimbrial outer membrane usher protein (PMI1466), flagellin protein (PMI1619), and regulator of swarming behavior (rsbA), which are responsible for adhesion, motility, and quorum sensing, respectively. Crystal violet assay, SEM, and confocal imaging confirmed the effectiveness of the PmA2G02 as an anti-biofilm agent. Moreover, as verified by qPCR, the expression levels of fimD, fliC2, and rsbA were significantly reduced compared to the untreated group. This study suggests that aptamer may be a potential alternative to traditional antibiotics for the treatment of CAUTIs caused by P. mirabilis. These findings shed light on the mechanisms by which the aptamer inhibits biofilm formation.
Collapse
Affiliation(s)
- Rajalakshmi Elumalai
- Molecular Genetics Laboratory, Department of Genetic Engineering, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Tamil Nadu, India
| | - Archana Vishwakarma
- Molecular Genetics Laboratory, Department of Genetic Engineering, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Tamil Nadu, India
| | - Anandkumar Balakrishnan
- Corrosion Science and Technology Division, Metallurgy and Materials Group, Indira Gandhi Centre for Atomic Research, Kalpakkam, India; Homi Bhabha National Institute Kalpakkam, Mumbai 400094, India
| | - Mohandass Ramya
- Molecular Genetics Laboratory, Department of Genetic Engineering, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Tamil Nadu, India.
| |
Collapse
|
117
|
Raj V, Raorane CJ, Shastri D, Kim SC, Lee S. Engineering a self-healing grafted chitosan-sodium alginate based hydrogel with potential keratinocyte cell migration property and inhibitory effect against fluconazole resistance Candida albicans biofilm. Int J Biol Macromol 2024; 261:129774. [PMID: 38286383 DOI: 10.1016/j.ijbiomac.2024.129774] [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: 11/02/2023] [Revised: 01/08/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024]
Abstract
Biofilms developed by microorganisms cause an extremely severe clinical problem that leads to drug failure. Bioactive polymeric hydrogels display potential for controlling the formation of microorganism-based biofilms, but their rapid biodegradability in these biofilm sites is still a major challenge. To overcome this, chitosan (CS), a natural functional biomaterial, has been used because of its effective penetrability in the cell wall of microorganisms; however, its fast biodegradability has restricted its further use. Hence, in this study, to improve the stability of CS and increase its penetration retention inside a biofilm, grafted CS was prepared and then crosslinked with sodium alginate (SA) to synthesize CS-poly(MA-co-AA)SA hydrogel via a free radical grafting method, therefore enhancing its antibiofilm efficiency against biofilms. The prepared hydrogel demonstrated excellent effectiveness against (≥90 % inhibition) biofilms of Candida albicans. Additionally, in vitro and in vivo safety assays established that the prepared hydrogel can be used in a biofilm microenvironment and might reduce drug resistance burden owing to its long-term antibiofilm effect and improved CS stability at the biofilm site. Furthermore, in vitro wound healing outcomes of hydrogel indicated its potential application for chronic wound treatment. This research opens a new advanced strategy for biofilm-associated infection treatment, including wound treatment.
Collapse
Affiliation(s)
- Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | | | - Divya Shastri
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea; College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Seong Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| |
Collapse
|
118
|
Li J, Yu Y, Zhou Y, Song J, Yang A, Wang M, Li Y, Wan M, Zhang C, Yang H, Bai Y, Wong WL, Pu H, Feng X. Multi-targeting oligopyridiniums: Rational design for biofilm dispersion and bacterial persister eradication. Bioorg Chem 2024; 144:107163. [PMID: 38306825 DOI: 10.1016/j.bioorg.2024.107163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/09/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
The development of effective antibacterial drugs to combat bacterial infections, particularly the biofilm-related infections, remains a challenge. There are two important features of bacterial biofilms, which are well-known critical factors causing biofilms hard-to-treat in clinical, including the dense and impermeable extracellular polymeric substances (EPS) and the metabolically repressed dormant and persistent bacterial population embedded. These characteristics largely increase the difficulty for regular antibiotic treatment due to insufficient penetration into EPS. In addition, the dormant bacteria are insensitive to the growth-inhibiting mechanism of traditional antibiotics. Herein, we explore the potential of a series of new oligopyridinium-based oligomers bearing a multi-biomacromolecule targeting function as the potent bacterial biofilm eradication agent. These oligomers were rationally designed to be "charge-on-backbone" that can offer a special alternating amphiphilicity. This novel and unique feature endows high affinity to bacterial membrane lipids, DNAs as well as proteins. Such a broad multi-targeting nature of molecules not only enables its penetration into EPS, but also plays vital roles in the bactericidal mechanism of action that is highly effective against dormant and persistent bacteria. Our in vitro, ex vivo, and in vivo studies demonstrated that OPc3, one of the most effective derivatives, was able to offer excellent antibacterial potency against a variety of bacteria and effectively eliminate biofilms in zebrafish models and mouse wound biofilm infection models.
Collapse
Affiliation(s)
- Jiaqi Li
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yue Yu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Anming Yang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Youzhi Li
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yugang Bai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong, China Hong Kong Special Administrative Region.
| | - Huangsheng Pu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China; College of Biology, Hunan University, Changsha, Hunan 410082, China; College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel Nano Optoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China; Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China.
| | - Xinxin Feng
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
119
|
Hussaini IM, Oyewole OA, Sulaiman MA, Dabban AI, Sulaiman AN, Tarek R. Microbial anti-biofilms: types and mechanism of action. Res Microbiol 2024; 175:104111. [PMID: 37844786 DOI: 10.1016/j.resmic.2023.104111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 10/18/2023]
Abstract
Biofilms have been recognized as a serious threat to public health as it protects microbes from antimicrobials, immune defence mechanisms, chemical treatments and nutritional stress. Biofilms are also a source of concern in industries and water treatment because their presence compromises the integrity of equipment. To overcome these problems, it is necessary to identify novel anti-biofilm compounds. Products of microorganisms have been identified as promising broad-spectrum anti-biofilm agents. These natural products include biosurfactants, antimicrobial peptides, enzymes and bioactive compounds. Anti-biofilm products of microbial origin are chemically diverse and possess a broad spectrum of activities against biofilms. The objective of this review is to give an overview of the different types of microbial anti-biofilm products and their mechanisms of action.
Collapse
Affiliation(s)
| | - Oluwafemi Adebayo Oyewole
- Department of Microbiology, School of Life Sciences, Federal University of Technology, Minna, Nigeria; African Center of Excellence for Mycotoxin and Food Safety, Federal University of Technology Minna, Nigeria.
| | | | | | - Asmau Nna Sulaiman
- Department of Microbiology, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Reham Tarek
- Department of Biotechnology, Cairo University, Egypt
| |
Collapse
|
120
|
Kaliniak S, Fiedoruk K, Spałek J, Piktel E, Durnaś B, Góźdź S, Bucki R, Okła S. Remodeling of Paranasal Sinuses Mucosa Functions in Response to Biofilm-Induced Inflammation. J Inflamm Res 2024; 17:1295-1323. [PMID: 38434581 PMCID: PMC10906676 DOI: 10.2147/jir.s443420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/23/2024] [Indexed: 03/05/2024] Open
Abstract
Rhinosinusitis (RS) is an acute (ARS) or chronic (CRS) inflammatory disease of the nasal and paranasal sinus mucosa. CRS is a heterogeneous condition characterized by distinct inflammatory patterns (endotypes) and phenotypes associated with the presence (CRSwNP) or absence (CRSsNP) of nasal polyps. Mucosal barrier and mucociliary clearance dysfunction, inflammatory cell infiltration, mucus hypersecretion, and tissue remodeling are the hallmarks of CRS. However, the underlying factors, their priority, and the mechanisms of inflammatory responses remain unclear. Several hypotheses have been proposed that link CRS etiology and pathogenesis with host (eg, "immune barrier") and exogenous factors (eg, bacterial/fungal pathogens, dysbiotic microbiota/biofilms, or staphylococcal superantigens). The abnormal interplay between these factors is likely central to the pathophysiology of CRS by triggering compensatory immune responses. Here, we discuss the role of the sinonasal microbiota in CRS and its biofilms in the context of mucosal zinc (Zn) deficiency, serving as a possible unifying link between five host and "bacterial" hypotheses of CRS that lead to sinus mucosa remodeling. To date, no clear correlation between sinonasal microbiota and CRS has been established. However, the predominance of Corynebacteria and Staphylococci and their interspecies relationships likely play a vital role in the formation of the CRS-associated microbiota. Zn-mediated "nutritional immunity", exerted via calprotectin, alongside the dysregulation of Zn-dependent cellular processes, could be a crucial microbiota-shaping factor in CRS. Similar to cystic fibrosis (CF), the role of SPLUNC1-mediated regulation of mucus volume and pH in CRS has been considered. We complement the biofilms' "mechanistic" and "mucin" hypotheses behind CRS pathogenesis with the "structural" one - associated with bacterial "corncob" structures. Finally, microbiota restoration approaches for CRS prevention and treatment are reviewed, including pre- and probiotics, as well as Nasal Microbiota Transplantation (NMT).
Collapse
Affiliation(s)
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Jakub Spałek
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Bonita Durnaś
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Stanisław Góźdź
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Sławomir Okła
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| |
Collapse
|
121
|
Xu LC, Booth JL, Lanza M, Ozdemir T, Huffer A, Chen C, Khursheed A, Sun D, Allcock HR, Siedlecki CA. In Vitro and In Vivo Assessment of the Infection Resistance and Biocompatibility of Small-Molecule-Modified Polyurethane Biomaterials. ACS APPLIED MATERIALS & INTERFACES 2024; 16:8474-8483. [PMID: 38330222 DOI: 10.1021/acsami.3c18231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Bacterial intracellular nucleotide second messenger signaling is involved in biofilm formation and regulates biofilm development. Interference with the bacterial nucleotide second messenger signaling provides a novel approach to control biofilm formation and limit microbial infection in medical devices. In this study, we tethered small-molecule derivatives of 4-arylazo-3,5-diamino-1H-pyrazole on polyurethane biomaterial surfaces and measured the biofilm resistance and initial biocompatibility of modified biomaterials in in vitro and in vivo settings. Results showed that small-molecule-modified surfaces significantly reduced the Staphylococcal epidermidis biofilm formation compared to unmodified surfaces and decreased the nucleotide levels of c-di-AMP in biofilm cells, suggesting that the tethered small molecules interfere with intracellular nucleotide signaling and inhibit biofilm formation. The hemocompatibility assay showed that the modified polyurethane films did not induce platelet activation or red blood cell hemolysis but significantly reduced plasma coagulation and platelet adhesion. The cytocompatibility assay with fibroblast cells showed that small-molecule-modified surfaces were noncytotoxic and cells appeared to be proliferating and growing on modified surfaces. In a 7-day subcutaneous infection rat model, the polymer samples were implanted in Wistar rats and inoculated with bacteria or PBS. Results show that modified polyurethane significantly reduced bacteria by ∼2.5 log units over unmodified films, and the modified polymers did not lead to additional irritation/toxicity to the animal tissues. Taken together, the results demonstrated that small molecules tethered on polymer surfaces remain active, and the modified polymers are biocompatible and resistant to microbial infection in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | - Tugba Ozdemir
- Department of Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota 57701, United States
| | - Amelia Huffer
- Department of Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota 57701, United States
| | - Chen Chen
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | | | | | - Harry R Allcock
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | | |
Collapse
|
122
|
El-Sherbiny GM, Farghal EE, Lila MK, Shetaia YM, Mohamed S, Elswify MMF. Antibiotic susceptibility and virulence factors of bacterial species among cancer patients. BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2024; 5:27-32. [PMID: 39416689 PMCID: PMC11446349 DOI: 10.1016/j.biotno.2024.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 10/19/2024]
Abstract
Antibiotic resistance is one of the most significant challenges of the 20-s century, and the misuse of antibiotics is a driver of antimicrobial resistance. This study aimed to assess the prevalence of multidrug resistance, and detection of its produce virulence factors, including extended-spectrum β-lactamases (ESβLs), biofilm, and siderophores produced by bacterial species isolated from cancer patients. One hundred and seventy-five Gram-negative bacterial isolates were isolated from different samples collected from cancer patients admitted to the National Cancer Institute (NCI), Cairo, Egypt, and processed by standard microbiological methods. One hundred and forty-three bacterial isolates were recovered from adult patients, and 32 were recovered from children. Escherichia coli showed the highest frequency (36%), followed by Klebsiella pneumonia (30.85%), Acinetobacter baummannii (14.28%), and Pseudomonas sp. (9.14%). Antibiotic profiles revealed that bacterial isolates are highly resistant to the most commonly available antibiotics. Amikacin and gentamicin were the most effective antibiotics against isolated Gram-negative bacteria. Moreover, the vast majority of bacterial stains produce virulence factors, including EsβLs, biofilm, and siderophores. E. coli isolates produced ESβLs with rates of 25.28%, Klebsiella pneumonia (11.0%), and Pseudomonas sp. (25.0%). Among these collected bacterial isolates, 132 (75.4%) have the ability to form a biofilm to different degrees. Also, the majority of the bacteria isolates generated siderophores, with 133 (75.94%). This study revealed that a significant distribution of multidrug-resistant pathogenic bacteria may increase the burden on healthcare to prevent infections in cancer patients.
Collapse
Affiliation(s)
- Gamal M. El-Sherbiny
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, 11884, Nasr City, Cairo, Egypt
| | - Eman E. Farghal
- Clinical and Chemical Pathology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | - Mohamed K. Lila
- Department of Microbiology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Yousseria M. Shetaia
- Department of Microbiology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - S.S. Mohamed
- Department of Microbiology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Marwa MF. Elswify
- Department Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
123
|
Mirzaei R, Campoccia D, Ravaioli S, Arciola CR. Emerging Issues and Initial Insights into Bacterial Biofilms: From Orthopedic Infection to Metabolomics. Antibiotics (Basel) 2024; 13:184. [PMID: 38391570 PMCID: PMC10885942 DOI: 10.3390/antibiotics13020184] [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: 12/19/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Bacterial biofilms, enigmatic communities of microorganisms enclosed in an extracellular matrix, still represent an open challenge in many clinical contexts, including orthopedics, where biofilm-associated bone and joint infections remain the main cause of implant failure. This study explores the scenario of biofilm infections, with a focus on those related to orthopedic implants, highlighting recently emerged substantial aspects of the pathogenesis and their potential repercussions on the clinic, as well as the progress and gaps that still exist in the diagnostics and management of these infections. The classic mechanisms through which biofilms form and the more recently proposed new ones are depicted. The ways in which bacteria hide, become impenetrable to antibiotics, and evade the immune defenses, creating reservoirs of bacteria difficult to detect and reach, are delineated, such as bacterial dormancy within biofilms, entry into host cells, and penetration into bone canaliculi. New findings on biofilm formation with host components are presented. The article also delves into the emerging and critical concept of immunometabolism, a key function of immune cells that biofilm interferes with. The growing potential of biofilm metabolomics in the diagnosis and therapy of biofilm infections is highlighted, referring to the latest research.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| |
Collapse
|
124
|
De K, Dey R, Acharya Y, Aswal VK, Haldar J. Cleavable Amphiphilic Biocides with Ester-Bearing Moieties: Aggregation Properties and Antibacterial Activity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 38324708 DOI: 10.1021/acs.langmuir.3c02771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The rise of multidrug-resistant bacterial infections and the dwindling supply of newly approved antibiotics have emerged as a grave threat to public health. Toward the ever-growing necessity of the development of novel antimicrobial agents, herein, we synthesized a series of cationic amphiphilic biocides featuring two cationic headgroups separated by different hydrophobic spacers, accompanied by the inclusion of two lipophilic tails through cleavable ester functionality. The detailed aggregation properties offered by these biocides were investigated by small-angle neutron scattering (SANS) and conductivity. The critical micellar concentration of the biocides and the size and shape of the micellar aggregates differed with variation of pendant and spacer hydrophobicity. Furthermore, the aggregation number and size of the micelles were found to vary with changing concentration and temperature. These easily synthesized biocides exhibited potent antibacterial properties against various multidrug-resistant bacteria. The optimized biocides with minimum hematotoxicity and potent antibacterial activity against methicillin-resistant Staphylococcus aureus and Acinetobacter baumannii exhibited rapid killing kinetics against planktonic bacteria. Also, these membrane-active agents were able to eradicate preformed biofilms. The enzymatic and acidic degradation profile further offered proof of gradual degradation. Collectively, these cleavable amphiphilic biocides demonstrated excellent potency for combating the multidrug-resistant bacterial infection.
Collapse
Affiliation(s)
- Kathakali De
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Rajib Dey
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Vinod Kumar Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre (BARC), Mumbai 400085, Maharashtra, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
125
|
Phucharoenrak P, Trachootham D. Bergaptol, a Major Furocoumarin in Citrus: Pharmacological Properties and Toxicity. Molecules 2024; 29:713. [PMID: 38338457 PMCID: PMC10856120 DOI: 10.3390/molecules29030713] [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: 12/31/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Bergaptol (5-hydroxypsoralen or 5-hydroxyfuranocoumarin) is a naturally occurring furanocoumarin widely found in citrus fruits, which has multiple health benefits. Nonetheless, no specific review articles on bergaptol have been published. Compiling updated information on bergaptol is crucial in guiding future research direction and application. The present review focuses on the research evidence related to the pharmacological properties and toxicity of bergaptol. Bergaptol has anti-inflammatory, antioxidant, anti-cancer, anti-osteoporosis, anti-microbial, and anti-lipidemic effects. It can inhibit the activities of cytochrome P450s (CYP), especially CYP2C9 and CYP3A4, thereby affecting the metabolism and concentrations of some drugs and toxins. Compared with other coumarins, bergaptol has the least potency to inhibit CYP3A4 in cancer cells. Instead, it can suppress drug efflux transporters, such as P-glycoprotein, thereby overcoming chemotherapeutic drug resistance. Furthermore, bergaptol has antimicrobial effects with a high potential for inhibition of quorum sensing. In vivo, bergaptol can be retained in plasma for longer than other coumarins. Nevertheless, its toxicity has not been clearly reported. In vitro study suggests that, unlike most furocoumarins, bergaptol is not phototoxic or photomutagenic. Existing research on bergaptol has mostly been conducted in vitro. Further in vivo and clinical studies are warranted to identify the safe and effective doses of bergaptol for its multimodal application.
Collapse
|
126
|
Tan Y, Zhao K, Yang S, Chen S, Li C, Han X, Li J, Hu K, Liu S, Ma M, Yu X, Zou L. Insights into antibiotic and heavy metal resistance interactions in Escherichia coli isolated from livestock manure and fertilized soil. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 351:119935. [PMID: 38154221 DOI: 10.1016/j.jenvman.2023.119935] [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: 08/29/2023] [Revised: 11/12/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
Heavy metal and antibiotic-resistant bacteria from livestock feces are ecological and public health problems. However, the distribution and relationships of antibiotic resistance genes (ARGs), heavy metal resistance genes (HMRGs), and virulence factors (VFs) and their transmission mechanisms remain unclear. Therefore, we investigated the resistance of Escherichia coli, the prevalence of its ARGs, HMRGs, and VFs, and their transmission mechanisms in livestock fresh feces (FF), composted feces (CF), and fertilized soil (FS). In total, 99.54% (n = 221) and 91.44% (n = 203) of E. coli were resistant to at least one antibiotic and one heavy metal, respectively. Additionally, 72.52% (n = 161) were multi-drug resistant (MDR), of which Cu-resistant E. coli accounted for 72.67% (117/161). More than 99.34% (88/89) of E. coli carried multidrug ARGs, VFs, and the Cu resistance genes cueO and cusABCRFS. The Cu resistance genes cueO and cusABCRFS were mainly located on chromosomes, and cueO and cusF were positively associated with HMRGs, ARGs, and VFs. The Cu resistance genes pcoABCDRS were located on the plasmid pLKYL-P02 flanked by ARGs in PF18C from FF group and on chromosomes flanked by HMRGs in SAXZ1-1 from FS group. These results improved our understanding of bacterial multidrug and heavy metal resistance in the environment.
Collapse
Affiliation(s)
- Yulan Tan
- College of Resource, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Ke Zhao
- College of Resource, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Shengzhi Yang
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China.
| | - Shujuan Chen
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China.
| | - Chun Li
- Sichuan Province Center for Animal Disease Prevention and Control, Chengdu, Sichuan, China.
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Jianlong Li
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China.
| | - Kaidi Hu
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China.
| | - Shuliang Liu
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China.
| | - Menggen Ma
- College of Resource, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Xiumei Yu
- College of Resource, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Likou Zou
- College of Resource, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
127
|
Bharathi D, Lee JH, Lee J. Enhancement of antimicrobial and antibiofilm activities of liposomal fatty acids. Colloids Surf B Biointerfaces 2024; 234:113698. [PMID: 38070368 DOI: 10.1016/j.colsurfb.2023.113698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 02/09/2024]
Abstract
Microbial biofilms are protected surface-attached communities of bacteria or fungi with high drug tolerance that typically cause persistent infections. Smart drug carriers are being explored as a promising platform of antimicrobials to address their recalcitrance to antibiotic agents and minimize the side effects of current therapies. In this study, soy lecithin liposomes loaded with lauric acid (LA) and myristoleic acid (MA) were formulated using an emulsification method, and their antibiofilm properties were evaluated. The physio-chemical properties of the most potent liposome were characterized using a zeta sizer, transmission electron microscopy (TEM), fourier transform infrared spectroscopy, and nuclear magnetic resonance spectroscopy. TEM and zeta sizer analysis of the liposome revealed a homogeneous spherical structure with an average size of 159.2 nm and zeta potential of - 5.4 mV. The unilamellar liposomes loaded with LA at 0.1-0.5 µg/mL achieved obvious antibiofilm efficiency against Staphylococcus aureus and Candida albicans and their dual biofilms. Also, LA-loaded liposome formulation efficiently disrupted preformed biofilms of S. aureus and C. albicans. Furthermore, formulated liposomal LA (0.1 µg/mL) exhibited 100-fold increased dual biofilm inhibition compared to LA alone. The single biofilms and dual biofilm formation on polystyrene were reduced as determined by 3D-bright field and scanning electron microscopy. Zeta potential measurements exhibited neutralized surface charge of S. aureus, and the liposomes inhibited hyphae formation in C. albicans. These findings demonstrated that the LA-incorporated liposomes have great potential to become a new, effective, and good antibiofilm agent for treating S. aureus and C. albicans infections.
Collapse
Affiliation(s)
- Devaraj Bharathi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
128
|
Song Q, Gao B, Zhang S, Hu C. Adopting the "Missile boats-Aircraft carrier" strategy via human-contact friendly oxidized starch to achieve rapid-sustainably antibacterial paperboards. Int J Biol Macromol 2024; 259:129066. [PMID: 38158062 DOI: 10.1016/j.ijbiomac.2023.129066] [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: 09/11/2023] [Revised: 12/14/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
Polysaccharide-based antibacterial agents have received tremendous attention for the facile fabrication, low toxicity, and high compatibility with carbohydrate polymers. However, the antimicrobial mechanism, activity, and cytotoxicity for human-contact paperboards of oxidized starch (OST) with high carboxyl content, has not been explored. Herein, OST-27- 75 with 27- 75 wt% carboxyl contents were fabricated by H2O2 and coated on paperboards. Strikingly, OST-55 coating layer (16 g/m2) did not exfoliate from paperboard and possessed the rapid-sustainable antibacterial performance against Staphylococcus aureus and Escherichia coli. The soluble and insoluble components of OST-55 (OST55-S: OST55-IS mass ratio = 1: 2.1) presented different antimicrobial features and herein they were characterized by GC-MS, FT-IR, H-NMR, XRD, bacteriostatic activities, biofilm formation inhibition and intracellular constituent leakage to survey the antibacterial mechanism. The results revealed OST55-S displayed an amorphous structure and possessed superior antibacterial activity against S. aureus (MIC = 4 mg/mL) and E. coli (MIC = 8 mg/mL). Distinctively, OST55-S could rapidly ionize [H+] like "missile boats" from small molecule saccharides, while OST55-IS polyelectrolyte could continuously and slowly release for [H+] like an "aircraft carrier" to inhibit biofilm formation and disrupt cell structure. Eventually, the "Missile boats-Aircraft carrier" strategy provided a green methodology to fabricate polymeric antibacterial agents and expanded the use of cellulose-based materials.
Collapse
Affiliation(s)
- Qiaowei Song
- Packaging Engineering Institute, Jinan University, Qianshan Road 206, Zhuhai, Guangdong 519070, China
| | - Bingbing Gao
- School of Mechanical and Automotive Engineering, South China University of Technology, No. 381 Wushan Road, Tianhe District, Guangzhou 510640, China
| | - Shuidong Zhang
- School of Mechanical and Automotive Engineering, South China University of Technology, No. 381 Wushan Road, Tianhe District, Guangzhou 510640, China; State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Changying Hu
- Packaging Engineering Institute, Jinan University, Qianshan Road 206, Zhuhai, Guangdong 519070, China; Department of Food Science & Engineering, Jinan University, Huangpu West Avenue 601, Guangzhou City 510632, Guangdong, China..
| |
Collapse
|
129
|
Nie M, Alejandro Valdes-Pena M, Frohock BH, Smits E, Daiker JC, Gilbertie JM, Schnabel LV, Pierce JG. Expanded library of novel 2,3-pyrrolidinedione analogues exhibit anti-biofilm activity. Bioorg Med Chem Lett 2024; 99:129609. [PMID: 38191097 PMCID: PMC10872213 DOI: 10.1016/j.bmcl.2024.129609] [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: 10/21/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Herein we report a new library of 2,3-pyrrolidinedione analogues that expands on our previous report on the antimicrobial studies of this heterocyclic scaffold. The novel 2,3-pyrrolidinediones reported herein have been evaluated against S. aureus and methicillin-resistant S. aureus (MRSA) biofilms, and this work constitutes our first report on the antibiofilm properties of this class of compounds. The antibiofilm activity of these 2,3-pyrrolidinediones has been assessed through minimum biofilm eradication concentration (MBEC) and minimum biofilm inhibition concentration (MBIC) assays. The compounds displayed antibiofilm properties and represent intriguing scaffolds for further optimization and development.
Collapse
Affiliation(s)
- Minhua Nie
- Department of Chemistry, College of Sciences, NC State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - M Alejandro Valdes-Pena
- Department of Chemistry, College of Sciences, NC State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - Bram H Frohock
- Department of Chemistry, College of Sciences, NC State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - Emma Smits
- Department of Chemistry, College of Sciences, NC State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - Jennifer C Daiker
- Department of Chemistry, College of Sciences, NC State University, Raleigh, NC 27695, USA; Department of Clinical Sciences College of Veterinary Medicine, NC State University, 1060 William Moore Drive, Raleigh, NC 27607, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - Jessica M Gilbertie
- Department of Clinical Sciences College of Veterinary Medicine, NC State University, 1060 William Moore Drive, Raleigh, NC 27607, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - Lauren V Schnabel
- Department of Clinical Sciences College of Veterinary Medicine, NC State University, 1060 William Moore Drive, Raleigh, NC 27607, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA
| | - Joshua G Pierce
- Department of Chemistry, College of Sciences, NC State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, NC State University, Raleigh, NC 27695, USA.
| |
Collapse
|
130
|
Merlani M, Nadaraia N, Barbakadze N, Amiranashvili L, Kakhabrishvili M, Petrou A, Carević T, Glamočlija J, Geronikaki A. Steroidal hydrazones as antimicrobial agents: biological evaluation and molecular docking studies. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:137-155. [PMID: 38312087 DOI: 10.1080/1062936x.2024.2309183] [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: 11/28/2023] [Accepted: 01/17/2024] [Indexed: 02/06/2024]
Abstract
Most of pharmaceutical agents display several or even many biological activities. It is obvious that testing even one compound for thousands of biological activities is a practically not reasonable task. Therefore, computer-aided prediction is the method of choice for the selection of the most promising bioassays for particular compounds. Using PASS Online software, we determined the probable antimicrobial activity of the 31 steroid derivatives. Experimental testing of the antimicrobial activity of the tested compounds by microdilution method confirmed the computational predictions. Furthermore, P. aeruginosa and C. albicans biofilm formation was investigated. Compound 11 showed a biofilm reduction by 42.26% at the MIC of the tested compound. The percentages are lower than ketoconazole, but very close to its activity.
Collapse
Affiliation(s)
- M Merlani
- TSMU I, Kutateladze Institute of Pharmacochemistry, Tbilisi, Georgia
| | - N Nadaraia
- TSMU I, Kutateladze Institute of Pharmacochemistry, Tbilisi, Georgia
| | - N Barbakadze
- TSMU I, Kutateladze Institute of Pharmacochemistry, Tbilisi, Georgia
| | - L Amiranashvili
- TSMU I, Kutateladze Institute of Pharmacochemistry, Tbilisi, Georgia
| | - M Kakhabrishvili
- TSMU I, Kutateladze Institute of Pharmacochemistry, Tbilisi, Georgia
| | - A Petrou
- School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - T Carević
- Department of Plant Physiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - J Glamočlija
- Department of Plant Physiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - A Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
131
|
Hayles A, Bright R, Nguyen NH, Truong VK, Vongsvivut J, Wood J, Kidd SP, Vasilev K. Staphylococcus aureus surface attachment selectively influences tolerance against charged antibiotics. Acta Biomater 2024; 175:369-381. [PMID: 38141932 DOI: 10.1016/j.actbio.2023.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/21/2023] [Accepted: 12/17/2023] [Indexed: 12/25/2023]
Abstract
The threat of infection during implant placement surgery remains a considerable burden for millions of patients worldwide. To combat this threat, clinicians employ a range of anti-infective strategies and practices. One of the most common interventions is the use of prophylactic antibiotic treatment during implant placement surgery. However, these practices can be detrimental by promoting the resilience of biofilm-forming bacteria and enabling them to persist throughout treatment and re-emerge later, causing a life-threatening infection. Thus, it is of the utmost importance to elucidate the events occurring during the initial stages of bacterial surface attachment and determine whether any biological processes may be targeted to improve surgical outcomes. Using gene expression analysis, we identified a cellular mechanism of S. aureus which modifies its cell surface charge following attachment to a medical grade titanium surface. We determined the upregulation of two systems involved in the d-alanylation of teichoic acids and the lysylation of phosphatidylglycerol. We supported these molecular findings by utilizing synchrotron-sourced attenuated total reflection Fourier-transform infrared microspectroscopy to analyze the biomolecular properties of the S. aureus cell surface following attachment. As a direct consequence, S. aureus quickly becomes substantially more tolerant to the positively charged vancomycin, but not the negatively charged cefazolin. The present study can assist clinicians in rationally selecting the most potent antibiotic in prophylaxis treatments. Furthermore, it highlights a cellular process that could potentially be targeted by novel technologies and strategies to improve the outcome of antibiotic prophylaxis during implant placement surgery. STATEMENT OF SIGNIFICANCE: The antibiotic tolerance of bacteria in biofilm is a well-established phenomenon. However, the physiological adaptations employed by Staphylococcus aureus to increase its antibiotic tolerance during the early stages of surface attachment are poorly understood. Using multiple techniques, including gene expression analysis and synchrotron-sourced Fourier-transform infrared microspectroscopy, we generated insights into the physiological response of S. aureus following attachment to a medical grade titanium surface. We showed that this phenotypic transition enables S. aureus to better tolerate the positively charged vancomycin, but not the negatively charged cefazolin. These findings shed light on the antibiotic tolerance mechanisms employed by S. aureus to survive prophylactically administered antibiotics and can help clinicians to protect patients from infections.
Collapse
Affiliation(s)
- Andrew Hayles
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042 Australia.
| | - Richard Bright
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042 Australia
| | - Ngoc Huu Nguyen
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, Australia
| | - Vi Khanh Truong
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042 Australia
| | - Jitraporn Vongsvivut
- Infrared Microspectroscopy (IRM) Beamline, ANSTO ‒ Australian Synchrotron, 800 Blackburn Road, Clayton, Victoria 3168, Australia
| | - Jonathan Wood
- Academic Unit of STEM, University of South Australia, Adelaide 5095, South Australia, Australia
| | - Stephen P Kidd
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia; Australian Centre for Antimicrobial Resistance Ecology, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Krasimir Vasilev
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042 Australia.
| |
Collapse
|
132
|
Camba C, Walter-Lakes B, Digal P, Taheri-Araghi S, Bezryadina A. Biofilm formation and manipulation with optical tweezers. BIOMEDICAL OPTICS EXPRESS 2024; 15:1181-1191. [PMID: 38404331 PMCID: PMC10890877 DOI: 10.1364/boe.510836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/23/2023] [Accepted: 01/05/2024] [Indexed: 02/27/2024]
Abstract
Some bacterial species form biofilms in suboptimal growth and environmental conditions. Biofilm structures allow the cells not only to optimize growth with nutrient availability but also to defend each other against external stress, such as antibiotics. Medical and bioengineering implications of biofilms have led to an increased interest in the regulation of bacterial biofilm formation. Prior research has primarily focused on mechanical and chemical approaches for stimulating and controlling biofilm formation, yet optical techniques are still largely unexplored. In this paper, we investigate the biofilm formation of Bacillus subtilis in a minimum biofilm-promoting medium (MSgg media) and explore the potential of optical trapping in regulating bacterial aggregation and biofilm development. Specifically, we determine the most advantageous stage of bacterial biofilm formation for optical manipulation and investigate the impact of optical trapping at different wavelengths on the aggregation of bacterial cells and the formation of biofilm. The investigation of optically regulated biofilm formation with optical tweezers presents innovative methodologies for the stimulation and suppression of biofilm growth through the application of lasers.
Collapse
Affiliation(s)
- Czarlyn Camba
- Department of Physics and Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, CA 91330, USA
| | - Brooke Walter-Lakes
- Department of Physics and Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, CA 91330, USA
| | - Phillip Digal
- Department of Physics and Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, CA 91330, USA
| | - Sattar Taheri-Araghi
- Department of Physics and Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, CA 91330, USA
| | - Anna Bezryadina
- Department of Physics and Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, CA 91330, USA
| |
Collapse
|
133
|
Ul Haq I, Khan TA, Krukiewicz K. Etiology, pathology, and host-impaired immunity in medical implant-associated infections. J Infect Public Health 2024; 17:189-203. [PMID: 38113816 DOI: 10.1016/j.jiph.2023.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/20/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Host impaired immunity and pathogens adhesion factors are the key elements in analyzing medical implant-associated infections (MIAI). The infection chances are further influenced by surface properties of implants. This review addresses the medical implant-associated pathogens and summarizes the etiology, pathology, and host-impaired immunity in MIAI. Several bacterial and fungal pathogens have been isolated from MIAI; together, they form cross-kingdom species biofilms and support each other in different ways. The adhesion factors initiate the pathogen's adherence on the implant's surface; however, implant-induced impaired immunity promotes the pathogen's colonization and biofilm formation. Depending on the implant's surface properties, immune cell functions get slow or get exaggerated and cause immunity-induced secondary complications resulting in resistant depression and immuno-incompetent fibro-inflammatory zone that compromise implant's performance. Such consequences lead to the unavoidable and straightforward conclusion for the downstream transformation of new ideas, such as the development of multifunctional implant coatings.
Collapse
Affiliation(s)
- Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland; Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil.
| | - Taj Ali Khan
- Division of Infectious Diseases & Global Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States; Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan.
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland.
| |
Collapse
|
134
|
Muzychka LV, Humeniuk NI, Boiko IO, Vrynchanu NO, Smolii OB. Synthesis and in vitro evaluation of antibacterial and antibiofilm activities of novel triphenylphosphonium-functionalized substituted pyrimidines. Chem Biol Drug Des 2024; 103:e14483. [PMID: 38355145 DOI: 10.1111/cbdd.14483] [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: 10/02/2023] [Revised: 01/19/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024]
Abstract
The increase in the prevalence of antibiotic-resistant pathogens leads to a decrease in the number of antimicrobial agents for the treatment of infections and prompts researchers to search for new effective antimicrobial drugs. This study reports the synthesis of novel triphenylphosphonium-functionalized substituted pyrimidines and in vitro evaluation of their antibacterial and antibiofilm activity. Most of the synthesized derivatives showed high antibacterial activity (MIC = 0.39-1.56 μg/mL) against the methicillin-resistant strain of S. aureus 222. Compounds 2a and 11 exhibited a high level of antibiofilm activity against S. aureus 222 and E. coli 311. The triphenylphosphonium-containing pyrimidines 11 and 2a reduced S. aureus 222 biofilm formation by 99.1% and 95.8%, respectively. In addition, compound 2a was the most active against E. coli 311 biofilm formation (the biomass decreased by 98.4%).
Collapse
Affiliation(s)
- Liubov V Muzychka
- Department of Chemistry of Natural Compounds, V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, NAS of Ukraine, Kyiv, Ukraine
| | - Nataliia I Humeniuk
- Laboratory of Pharmacology of Antimicrobials, SI Institute of Pharmacology and Toxicology, NAMS of Ukraine, Kyiv, Ukraine
| | - Iryna O Boiko
- Laboratory of Pharmacology of Antimicrobials, SI Institute of Pharmacology and Toxicology, NAMS of Ukraine, Kyiv, Ukraine
| | - Nina O Vrynchanu
- Laboratory of Pharmacology of Antimicrobials, SI Institute of Pharmacology and Toxicology, NAMS of Ukraine, Kyiv, Ukraine
| | - Oleg B Smolii
- Department of Chemistry of Natural Compounds, V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
135
|
El-Sawy YNA, Abdel-Salam AB, Abd-Elhady HM, Abou-Taleb KAA, Ahmed RF. Elimination of detached Listeria monocytogenes from the biofilm on stainless steel surfaces during milk and cheese processing using natural plant extracts. Sci Rep 2024; 14:2288. [PMID: 38280925 PMCID: PMC10821901 DOI: 10.1038/s41598-024-52394-9] [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: 08/12/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024] Open
Abstract
Bacterial cells can form biofilm on food contact surfaces, becoming a source of food contamination with profound health implications. The current study aimed to determine some Egyptian medicinal plants antibacterial and antibiofilm effects against foodborne bacterial strains in milk plants. Results indicated that four ethanolic plant extracts, Cinnamon (Cinnamomum verum), Chamomile (Matricaria chamomilla), Marigold (Calendula officinalis), and Sage (Salvia officinalis), had antibacterial (12.0-26.5 mm of inhibition zone diameter) and antibiofilm (10-99%) activities against Staphylococcus aureus, Bacillus cereus, Listeria monocytogenes and Salmonella Typhimurium. The tested extracts had minimum inhibitory concentration values between 0.14 and 2.50 mg/ml and minimum bactericidal concentration values between 0.14 and 12.50 mg/ml. L. monocytogenes was more sensitive for all tested ethanolic extracts; Sage and Cinnamon showed a bacteriocidal effect, while Chamomile and Marigold were bacteriostatic. The ethanolic extracts mixture from Chamomile, Sage, and Cinnamon was chosen for its antibiofilm activity against L. monocytogenes using L-optimal mixture design. Gas chromatography and mass spectrometry analysis showed that this mixture contained 12 chemical compounds, where 2-Propenal,3-phenyl- had the maximum area % (34.82%). At concentrations up to 500 µg/ml, it had no cytotoxicity in the normal Vero cell line, and the IC50 value was 671.76 ± 9.03 µg/ml. Also, this mixture showed the most significant antibacterial effect against detached L. monocytogenes cells from formed biofilm in stainless steel milk tanks. At the same time, white soft cheese fortified with this mixture was significantly accepted overall for the panelist (92.2 ± 2.7) than other cheese samples, including the control group.
Collapse
Affiliation(s)
- Yasmine N A El-Sawy
- Agricultural Microbiology Department, Faculty of Agriculture, Ain Shams University, Hadayek Shubra, Cairo, 11241, Egypt
| | - Ayah B Abdel-Salam
- Food Hygiene and Control Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Hemmat M Abd-Elhady
- Agricultural Microbiology Department, Faculty of Agriculture, Ain Shams University, Hadayek Shubra, Cairo, 11241, Egypt
| | - Khadiga A A Abou-Taleb
- Agricultural Microbiology Department, Faculty of Agriculture, Ain Shams University, Hadayek Shubra, Cairo, 11241, Egypt.
| | - Rania F Ahmed
- Agricultural Microbiology Department, Faculty of Agriculture, Ain Shams University, Hadayek Shubra, Cairo, 11241, Egypt
| |
Collapse
|
136
|
Tamilselvi B, Bhuvaneshwari DS, Karuppasamy P, Padmavathy S, Nikhil S, Siddegowda SB, Ananda Murthy HC. Investigation of Corrosion Inhibition of Mild Steel in 0.5 M H 2SO 4 with Lachancea fermentati Inhibitor Extracted from Rotten Grapefruits ( Vitis vinifera): Adsorption, Thermodynamic, Electrochemical, and Quantum Chemical Studies. ACS PHYSICAL CHEMISTRY AU 2024; 4:67-84. [PMID: 38283783 PMCID: PMC10811774 DOI: 10.1021/acsphyschemau.3c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 01/30/2024]
Abstract
Corrosion inhibition of mild steel (MS) was studied using Lachancea fermentati isolate in 0.5 M H2SO4, which was isolated from rotten grapes (Vitis vinifera) via biofilm formation. Biofilm over the MS surface was asserted by employing FT-IR and FE-SEM with EDXS, electrochemical impedance spectroscopy (EIS), AFM, and DFT-ESP techniques. The weight loss experiments and temperature studies supported the physical adsorption behavior of the corrosion inhibitors. The maximum inhibition efficiency (IE) value (90%) was observed at 293 K for 9 × 106 cfu/mL of Lachancea fermentati isolate. The adsorption of Lachancea fermentati isolate on the surface of MS confirms Langmuir's adsorption isotherm model, and the -ΔG values indicate the spontaneous adsorption of inhibitor over the MS surface. Electrochemical studies, such as potentiodynamic polarization (PDP) and EIS were carried out to investigate the charge transfer (CT) reaction of the Lachancea fermentati isolate. Tafel polarization curves reveal that the Lachancea fermentati isolate acts as a mixed type of inhibitor. The Nyquist plots (EIS) indicate the increase in charge transfer resistance (Rct) and decrease of double-layer capacitance (Cdl) values when increasing the concentration of Lachancea fermentati isolate. The spectral studies, such as UV-vis and FT-IR, confirm the formation of a complex between MS and the Lachancea fermentati isolate inhibitor. The formation of biofilm on the MS surface was confirmed by FE-SEM, EDXS, and XPS analysis. The proposed bioinhibitor shows great potential for the corrosion inhibition of mild steel in acid media.
Collapse
Affiliation(s)
- Baluchamy Tamilselvi
- Department
of Chemistry, Thiagarajar College, Madurai 625009, Tamil Nadu, India
- Department
of Chemistry, K.L.N. College of Engineering, Pottapalaiyam 630612, Tamil Nadu, India
| | | | | | - Sethuramasamy Padmavathy
- Department
of Microbiology and Biotechnology, Thiagarajar
College, Madurai 625009, Tamil Nadu, India
| | - Santhosh Nikhil
- School
of Chemistry, Madurai Kamaraj University, Madurai 625009, Tamil Nadu, India
| | | | - H C Ananda Murthy
- Department
of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888 Adama, Ethiopia
- Department
of Prosthodontics, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Science
(SIMATS), Saveetha University, Chennai 600077, Tamil
Nadu, India
| |
Collapse
|
137
|
Le KCM, Tran ATQ, Vu MP, Duong PVQ, Nguyen KT. Preventing Static Biofilm Formation of Staphylococcus aureus on Different Types of Surfaces Using Microbubbles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1698-1706. [PMID: 38198688 DOI: 10.1021/acs.langmuir.3c02773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Bacterial fouling and biofilm formation on surfaces have been ongoing problems in real life as well as in the medical field. Different approaches have been taken to tackle the issues, from costly surface modification to antibiotic-delivering strategies. In this study, we examined the potential of using stabilized microbubbles (MBs) to shield against bacterial adhesion. Three types of surfaces were tested: hydrophilic glass (hydrophilic surface), neutral hydrophobic polystyrene (PS)-coated surfaces, and negatively charged hydrophobic octadecyltrichlorosilane (OTS)-coated surfaces. By evaluating the colony-forming unit (CFU) values from each surface, MBs stabilized by 0.05 mM SDS were shown to only produce significant reduction of Staphylococcus aureus adhesion on PS surfaces, up to 60.29 and 82.32% compared to no-MB PS surfaces, and no-MB uncoated surfaces, correspondingly, due to the appropriate size, stability, and negative charges of the MB shielding layer. On the other hand, OTS coatings had an intrinsic antiadhesion effect (69.83% compared to uncoated surface), given that the negatively charged OTS-aqueous interface or surface porosity nature of the coating prohibited the attachment of MBs, leading to the elimination of the antifouling effect of MBs. Ultimately, MBs gave better shielding results than surface modification when compared to uncoated surfaces and hence can be applied more widely.
Collapse
Affiliation(s)
- Khoa C M Le
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Viet Nam
| | - Anh T Q Tran
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Viet Nam
| | - Mai P Vu
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Viet Nam
| | - Phuong V Q Duong
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Viet Nam
| | - Khoi T Nguyen
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Viet Nam
| |
Collapse
|
138
|
Hernández FLC, Virguez JV, Vesga JFG, Castellanos ML, Beltrán GR, Toquica LDL, Gomez CNS, Ríos MVS, Bolívar YRC, Sanchez JIA. Effect of COVID-19 on infections associated with medical devices in critical care. BMC Infect Dis 2024; 24:110. [PMID: 38254034 PMCID: PMC10801999 DOI: 10.1186/s12879-023-08934-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVES This study explores the hypothesis that COVID-19 patients are at a heightened risk of healthcare-associated infections (HAIs) associated with medical device usage compared to non-COVID-19 patients. Our primary objective was to investigate the correlation between COVID-19 infection in ICU patients and subsequent HAIs following invasive medical device insertion. Additionally, we aim to assess the impact of SARS-CoV-2 infection on onset times concerning specific microorganisms and the type of medical device, providing valuable insights into this intricate relationship in intensive care settings. METHODOLOGY A retrospective cohort study was conducted using ICU patient records at our hospital from 2020 to 2022. This investigation entailed evaluating the timing of HAIs while distinguishing between patients with and without SARS-CoV-2 infection. We identified and analyzed the type of isolation and infection attributed to the medical device while controlling for ICU duration and ventilator days using Cox regression. RESULTS Our study included 127 patients without SARS-CoV-2 infection and 140 patients with SARS-CoV-2 infection. The findings indicated a higher incidence of HAI caused by various microorganisms associated with any medical device in patients with SARS-CoV-2 (HR = 6.86; 95% CI-95%: 3.26-14.43; p < 0.01). After adjusting for ICU duration and ventilator days, a heightened frequency of HAIs persisted in SARS-CoV-2-infected individuals. However, a detailed examination of HAIs revealed that only ventilation-associated pneumonia (VAP) displayed a significant association (HR = 6.69; 95% CI: 2.59-17.31; p < 0.01). A statistically significant correlation between SARS-CoV-2 infection and the isolation of S. aureus was also observed (p = 0.034). The prevalence of S. aureus infection was notably higher in patients with SARS-CoV-2 (RR = 8.080; 95% CI: 1.052-62.068; p < 0.01). CONCLUSIONS The frequency of pathogen isolates in invasive medical devices exhibited an association with SARS-CoV-2 infection. Critically ill patients with SARS-CoV-2 are more prone to developing early-onset VAP than those without SARS-CoV-2 infection.
Collapse
|
139
|
Grooters KE, Ku JC, Richter DM, Krinock MJ, Minor A, Li P, Kim A, Sawyer R, Li Y. Strategies for combating antibiotic resistance in bacterial biofilms. Front Cell Infect Microbiol 2024; 14:1352273. [PMID: 38322672 PMCID: PMC10846525 DOI: 10.3389/fcimb.2024.1352273] [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: 12/07/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Biofilms, which are complexes of microorganisms that adhere to surfaces and secrete protective extracellular matrices, wield substantial influence across diverse domains such as medicine, industry, and environmental science. Despite ongoing challenges posed by biofilms in clinical medicine, research in this field remains dynamic and indeterminate. This article provides a contemporary assessment of biofilms and their treatment, with a focus on recent advances, to chronicle the evolving landscape of biofilm research.
Collapse
Affiliation(s)
- Kayla E. Grooters
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Jennifer C. Ku
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - David M. Richter
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Matthew J. Krinock
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Ashley Minor
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Patrick Li
- University of Michigan, Ann Arbor, MI, United States
- Division of Biomedical Engineering, Department of Orthopedic Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Audrey Kim
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Robert Sawyer
- Department of Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Yong Li
- Division of Biomedical Engineering, Department of Orthopedic Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
140
|
Somrani M, Huertas JP, Iguaz A, Debbabi H, Palop A. Biofilm busters: Exploring the antimicrobial and antibiofilm properties of essential oils against Salmonella Enteritidis. FOOD SCI TECHNOL INT 2024:10820132241227004. [PMID: 38234112 DOI: 10.1177/10820132241227004] [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: 01/19/2024]
Abstract
This study delves into an exploration of the antimicrobial and antibiofilm properties of the essential oils (EOs) of cinnamon, garlic, and onion on Salmonella Enteritidis. Firstly, disc diffusion and minimum inhibitory concentration (MIC) techniques were employed to assess the antibacterial activity of the EOs. Additionally, the study explored the effect of these EOs on both initial cell attachment and 24 h-preformed biofilms. The crystal violet assay was implemented to evaluate biofilm biomass. The findings revealed that cinnamon EO exhibited the highest anti-biofilm activity. Furthermore, initial cell attachment inhibition at MIC ranged between 50 and 65% for the three oils, while inhibition rates on preformed structures were lower than 40% for all EOs at this MIC concentration. The study also found that the effects of these oils were dosage- and time-dependent (p < 0.05), thereby urging the adoption of these natural extracts as effective strategies for combating Salmonella biofilms.
Collapse
Affiliation(s)
- Mariem Somrani
- Department of Agricultural Engineering, Institute of Plant Biotechnology, Universidad Politécnica de Cartagena, Murcia, Spain
- Department of AgriFood Industries, National Agronomic Institute of Tunisia, University of Carthage, Tunis, Tunisia
| | - Juan-Pablo Huertas
- Department of Agricultural Engineering, Institute of Plant Biotechnology, Universidad Politécnica de Cartagena, Murcia, Spain
| | - Asunción Iguaz
- Department of Agricultural Engineering, Institute of Plant Biotechnology, Universidad Politécnica de Cartagena, Murcia, Spain
| | - Hajer Debbabi
- Department of AgriFood Industries, National Agronomic Institute of Tunisia, University of Carthage, Tunis, Tunisia
| | - Alfredo Palop
- Department of Agricultural Engineering, Institute of Plant Biotechnology, Universidad Politécnica de Cartagena, Murcia, Spain
| |
Collapse
|
141
|
Bhattacharya M, Horswill AR. The role of human extracellular matrix proteins in defining Staphylococcus aureus biofilm infections. FEMS Microbiol Rev 2024; 48:fuae002. [PMID: 38337187 PMCID: PMC10873506 DOI: 10.1093/femsre/fuae002] [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: 07/19/2023] [Revised: 01/26/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
Twenty to forty one percent of the world's population is either transiently or permanently colonized by the Gram-positive bacterium, Staphylococcus aureus. In 2017, the CDC designated methicillin-resistant S. aureus (MRSA) as a serious threat, reporting ∼300 000 cases of MRSA-associated hospitalizations annually, resulting in over 19 000 deaths, surpassing that of HIV in the USA. S. aureus is a proficient biofilm-forming organism that rapidly acquires resistance to antibiotics, most commonly methicillin (MRSA). This review focuses on a large group of (>30) S. aureus adhesins, either surface-associated or secreted that are designed to specifically bind to 15 or more of the proteins that form key components of the human extracellular matrix (hECM). Importantly, this includes hECM proteins that are pivotal to the homeostasis of almost every tissue environment [collagen (skin), proteoglycans (lung), hemoglobin (blood), elastin, laminin, fibrinogen, fibronectin, and fibrin (multiple organs)]. These adhesins offer S. aureus the potential to establish an infection in every sterile tissue niche. These infections often endure repeated immune onslaught, developing into chronic, biofilm-associated conditions that are tolerant to ∼1000 times the clinically prescribed dose of antibiotics. Depending on the infection and the immune response, this allows S. aureus to seamlessly transition from colonizer to pathogen by subtly manipulating the host against itself while providing the time and stealth that it requires to establish and persist as a biofilm. This is a comprehensive discussion of the interaction between S. aureus biofilms and the hECM. We provide particular focus on the role of these interactions in pathogenesis and, consequently, the clinical implications for the prevention and treatment of S. aureus biofilm infections.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, CO 80045, United States
| |
Collapse
|
142
|
Gliźniewicz M, Miłek D, Olszewska P, Czajkowski A, Serwin N, Cecerska-Heryć E, Dołęgowska B, Grygorcewicz B. Advances in bacteriophage-mediated strategies for combating polymicrobial biofilms. Front Microbiol 2024; 14:1320345. [PMID: 38249486 PMCID: PMC10797108 DOI: 10.3389/fmicb.2023.1320345] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
Bacteria and fungi tend to coexist within biofilms instead of in planktonic states. Usually, such communities include cross-kingdom microorganisms, which make them harder to remove from abiotic surfaces or infection sites. Additionally, the produced biofilm matrix protects embedded microorganisms from antibiotics, disinfectants, or the host immune system. Therefore, classic therapies based on antibiotics might be ineffective, especially when multidrug-resistant bacteria are causative factors. The complexities surrounding the eradication of biofilms from diverse surfaces and the human body have spurred the exploration of alternative therapeutic modalities. Among these options, bacteriophages and their enzymatic counterparts have emerged as promising candidates, either employed independently or in synergy with antibiotics and other agents. Phages are natural bacteria killers because of mechanisms of action that differ from antibiotics, phages might answer worldwide problems with bacterial infections. In this review, we report the attempts to use bacteriophages in combating polymicrobial biofilms in in vitro studies, using different models, including the therapeutical use of phages. In addition, we sum up the advantages, disadvantages, and perspectives of phage therapy.
Collapse
Affiliation(s)
- Marta Gliźniewicz
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Dominika Miłek
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Patrycja Olszewska
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Artur Czajkowski
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Natalia Serwin
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Elżbieta Cecerska-Heryć
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Barbara Dołęgowska
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bartłomiej Grygorcewicz
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
- Department of Chemical Technology and Engineering, Institute of Chemical Engineering and Environmental Protection Processes, West Pomeranian University of Technology, Szczecin, Poland
| |
Collapse
|
143
|
Khosakueng M, Taweechaisupapong S, Boonyanugomol W, Prapatpong P, Wongkaewkhiaw S, Kanthawong S. Cymbopogon citratus L. essential oil as a potential anti-biofilm agent active against antibiotic-resistant bacteria isolated from chronic rhinosinusitis patients. BIOFOULING 2024; 40:26-39. [PMID: 38286789 DOI: 10.1080/08927014.2024.2305387] [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: 07/19/2023] [Accepted: 01/07/2024] [Indexed: 01/31/2024]
Abstract
Chronic rhinosinusitis (CRS) is long-term inflammation of the sinuses that can be caused by infection due to antibiotic-resistant bacteria. Biofilm developed by microbes is postulated to cause antibiotic treatment failure. Thus, the anti-biofilm activities of seven Thai herbal essential oils (EOs) against antibiotic-resistant bacteria isolated from CRS patients was investigated. Lemongrass (Cymbopogon citratus L.) EO showed the most effective antibiofilm activity against Klebsiella pneumoniae, Pseudomonas aeruginosa and Staphylococcus epidermidis grown as biofilm. GC-MS analysis found that myrcene was the major bioactive compound. Pretreatment with lemongrass EO significantly inhibited biofilm formation of all bacterial strains in more than 50% of cases. Furthermore, confocal microscopy analysis revealed the biofilm-disrupting activity of lemongrass EO against the biofilm matrix of all these bacterial species and also increased P. aeruginosa swarming motility with no toxicity to human cells. These results suggest that lemongrass EO has promising clinical applications as an anti-biofilm agent for CRS patients.
Collapse
Affiliation(s)
- Mintra Khosakueng
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Wongwarut Boonyanugomol
- Department of Medical Science, Amnatcharoen Campus, Mahidol University, Amnatcharoen, Thailand
| | - Pornpan Prapatpong
- Department of Public Health, Amnatcharoen Campus, Mahidol University, Amnatcharoen, Thailand
| | - Saharut Wongkaewkhiaw
- School of Dentistry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Sakawrat Kanthawong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
144
|
Sukmarini L, Atikana A, Hertiani T. Antibiofilm activity of marine microbial natural products: potential peptide- and polyketide-derived molecules from marine microbes toward targeting biofilm-forming pathogens. J Nat Med 2024; 78:1-20. [PMID: 37930514 DOI: 10.1007/s11418-023-01754-2] [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: 05/18/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023]
Abstract
Controlling and treating biofilm-related infections is challenging because of the widespread presence of multidrug-resistant microbes. Biofilm, a naturally occurring matrix of microbial aggregates, has developed intricate and diverse resistance mechanisms against many currently used antibiotics. This poses a significant problem, especially for human health, including clinically chronic infectious diseases. Thus, there is an urgent need to search for and develop new and more effective antibiotics. As the marine environment is recognized as a promising reservoir of new biologically active molecules with potential pharmacological properties, marine natural products, particularly those of microbial origin, have emerged as a promising source of antibiofilm agents. Marine microbes represent an untapped source of secondary metabolites with antimicrobial activity. Furthermore, marine natural products, owing to their self-defense mechanisms and adaptation to harsh conditions, encompass a wide range of chemical compounds, including peptides and polyketides, which are primarily found in microbes. These molecules can be exploited to provide novel and unique structures for developing alternative antibiotics as effective antibiofilm agents. This review focuses on the possible antibiofilm mechanism of these marine microbial molecules against biofilm-forming pathogens. It provides an overview of biofilm development, its recalcitrant mode of action, strategies for the development of antibiofilm agents, and their assessments. The review also revisits some selected peptides and polyketides from marine microbes reported between 2016 and 2023, highlighting their moderate and considerable antibiofilm activities. Moreover, their antibiofilm mechanisms, such as adhesion modulation/inhibition targeting biofilm-forming pathogens, quorum sensing intervention and inhibition, and extracellular polymeric substance disruption, are highlighted herein.
Collapse
Affiliation(s)
- Linda Sukmarini
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), KST Soekarno, Jl. Raya Jakarta-Bogor Km. 46, Cibinong, West Java, 16911, Indonesia.
- Indonesian Biofilm Research Collaboration Center, Jl. Farmako Sekip Utara, Yogyakarta, 55281, Indonesia.
| | - Akhirta Atikana
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), KST Soekarno, Jl. Raya Jakarta-Bogor Km. 46, Cibinong, West Java, 16911, Indonesia
- Indonesian Biofilm Research Collaboration Center, Jl. Farmako Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Triana Hertiani
- Indonesian Biofilm Research Collaboration Center, Jl. Farmako Sekip Utara, Yogyakarta, 55281, Indonesia.
- Pharmaceutical Biology Department, Faculty of Pharmacy, Gadjah Mada University, Jl. Sekip Utara, Yogyakarta, 55281, Indonesia.
| |
Collapse
|
145
|
Celebi O, Celebi D, Baser S, Aydın E, Rakıcı E, Uğraş S, Ağyar Yoldaş P, Baygutalp NK, Abd El-Aty AM. Antibacterial Activity of Boron Compounds Against Biofilm-Forming Pathogens. Biol Trace Elem Res 2024; 202:346-359. [PMID: 37464169 DOI: 10.1007/s12011-023-03768-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023]
Abstract
This study aimed to evaluate the antibacterial activity of nine boron derivatives against biofilm-forming pathogenic bacteria. The effect of boron derivatives (CMB, calcium metaborate; SMTB, sodium metaborate tetrahydrate; ZB, zinc borate; STFB, sodium tetra fluorine borate; STB, sodium tetraborate; PTFB, potassium tetra fluor borate; APTB, ammonium pentabo-rate tetrahydrate; SPM, sodium perborate monohydrate; Borax, ATFB, ammonium tetra fluorine borate) on bacteria isolated from blood culture was determined by the minimum inhibitory concentration (MIC) method. Then, biofilm formation potentials on microplates, tubes, and Congo red agar were examined. The cytotoxicity of boron derivatives was determined by using WST-1-based methods. The interaction between the biofilm-forming bacteria, fibroblast cells, and boron derivatives was determined with the infection model. We found that the sodium metaborate tetrahydrate molecule was effective against all pathogens. According to the optical density values detected at 630 nm in microplates, meticillin-resistant Staphylococcus aureus was observed to have the most substantial biofilm ability at 0.257 nm. As a result of cytotoxicity studies, it has been determined that a 1 µg/L concentration of boron derivatives is not toxic to fibroblast L929 cells. In cell culture experiments, these boron derivatives have very serious inhibitory activity against biofilm-forming pathogens in a short treatment period, such as 2-4 h. Furthermore, using these molecules on inanimate surfaces affected by biofilms would be appropriate instead of living cells.
Collapse
Affiliation(s)
- Ozgur Celebi
- Department of Medical Microbiology, Faculty of Medicine, Ataturk University, 25240, Erzurum, Turkey
| | - Demet Celebi
- Department of Microbiology, Faculty of Veterinary Medicine, Ataturk University, 25240, Erzurum, Turkey
- Ataturk University Vaccine Application and Development Center, Ataturk University, 25240, Erzurum, Turkey
| | - Sumeyye Baser
- Department of Medical Microbiology, Faculty of Medicine, Ataturk University, 25240, Erzurum, Turkey
| | - Elif Aydın
- Vocational School of Health Services, Kütahya Health Sciences University, Kütahya, Turkey.
| | - Erva Rakıcı
- Department of Medical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53020, Rize, Turkey
| | - Serpil Uğraş
- Department of Field Crops, Faculty of Agriculture and Natural Science, Duzce University, 81620, Düzce, Turkey
| | - Pınar Ağyar Yoldaş
- Traditional and Complementary Medicine Applied and Research Center, Duzce University, Duzce, Turkey
| | - Nurcan Kılıç Baygutalp
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, 25240, Erzurum, Turkey
| | - A M Abd El-Aty
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240, Erzurum, Turkey
| |
Collapse
|
146
|
Tang L, Zhang Z, Ding W, Tang J, Deng X, He Q, Kong X, Chen J, Truong TMH, Wang G, Zhu X, Ding W. Preparation, characterization, and Staphylococcus aureus biofilm elimination effect of baicalein-loaded tyrosine/hyaluronic acid/β-cyclodextrin-grafted chitosan nano-delivery system. Int J Biol Macromol 2024; 254:128066. [PMID: 37963503 DOI: 10.1016/j.ijbiomac.2023.128066] [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: 08/26/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 11/16/2023]
Abstract
Staphylococcus aureus (S. aureus) is an important cause of infections associated with implanted medical devices due to the formation of bacterial biofilm, which can prevent the penetration of drugs, thus posing a serious multi-drug resistance. Methicillin-resistant Staphylococcus aureus (MRSA) is one of them. In order to enhance the biofilm elimination effect of Baicalein (BA), a BA-loaded Tyr/HA/CD-CS nano-delivery system was successfully prepared using β-cyclodextrin grafted with chitosan (CD-CS), Hyaluronic Acid (HA), and D-Tyrosine (D-Tyr). The Tyr/HA/CD-CS-BA-NPs have a uniform particle size distribution with a particle size of 238.1 ± 3.06 nm and a PDI of 0.130 ± 0.02. The NPs showed an obvious inhibitory effect on planktonic bacteria with a MIC of 12.5 μg/mL. In vivo and in vitro tests showed that the NPs could enhance the elimination effect of BA on the MRSA biofilm. The results of Confocal Laser Scanning Microscopy (CLSM), Live & Dead Kit, and colony count experiments illustrated that Tyr/HA/CD-CS-BA-NPs could enhance the permeability of drugs to the biofilm and improve the ability to kill the biofilm bacteria, which may be an important mechanism to enhance the elimination of the MRSA biofilm. These findings will help develop new, effective medicaments for treating bacterial biofilm infections.
Collapse
Affiliation(s)
- Ling Tang
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | - Zhongbin Zhang
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China; Key Laboratory of Common Technology of Chinese Medicine Preparations, Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | - Wenyou Ding
- College of Basic Courses, Wuhan Donghu University, 430000, Hubei, China
| | - Jing Tang
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | - Xiuzhen Deng
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | - Qiumei He
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | - Xiangyu Kong
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | - Jinqing Chen
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China
| | | | - Gang Wang
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China; Key Laboratory of Common Technology of Chinese Medicine Preparations, Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China.
| | - Xiaoyong Zhu
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China; Key Laboratory of Common Technology of Chinese Medicine Preparations, Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China.
| | - Wenya Ding
- Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China; Key Laboratory of Common Technology of Chinese Medicine Preparations, Guangxi University of Chinese Medicine, Nanning 530022, Guangxi, China.
| |
Collapse
|
147
|
Aburayyan WS, Seder N, Al-fawares O, Fararjeh A, Majali IS, Al-Hajaya Y. Characterization of Antibiofilm and Antimicrobial Effects of Trigona Stingless Bee Honey Compared to Stinging Bee Centaurea hyalolepis and Citrus Honeys. J Evid Based Integr Med 2024; 29:2515690X241271978. [PMID: 39118572 PMCID: PMC11311187 DOI: 10.1177/2515690x241271978] [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: 06/11/2023] [Revised: 05/15/2024] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
The antibiofilm and antimicrobial properties of tropical honey types including Malaysian stingless bee honey remain explicitly unexplored when compared with Apies honey. The antibiofilm and antimicrobial activities of the Malaysian Trigona honey were characterized with two stinging bee honey types (Centaurea hyalolepis and Citrus honeys) from Jordan. The antibiofilm and antimicrobial investigations were conducted on a set of seven microbial strains; five bacterial species of Pseudomonas aeruginosa ATCC 10145, Streptococcus pyogenes ATCC 19615, Staphylococcus aureus ATCC 25923, Escherichia coli ATCC 25922, Klebsiella pneumoniae ATCC 13883, and two fungal strains Candida albicans ATCC 10231 and Candida krusei ATCC 14243. The antimicrobial investigations revealed a broad spectrum activity for Trigona honey against Gram-positive, Gram-negative, and fungal strains over the two honey types. One-way ANOVA showed a significant difference (p < 0.001) in the zone of inhibition ranging from 9 to 25 mm and minimum inhibition activity (MIC) ranged from 9.4-29.6% (w/v) against the microbial strains. Moreover, the addition of honey to established biofilms has induced a degradation activity in the biofilm mass. Two-way ANOVA showed a significant biofilm degradation proportion (p < 0.001) ranging from 1.3% to 91.3% following treatment with Trigona honey and the other honey types in relevance to the concentration ranging from 10% to 50% (w/v). Moreover, the antibiofilm activity was highly consistent with MIC affecting bacterial growth inhibition. In conclusion, a robust antimicrobial and antibiofilm activity for Trigona stingless bee honey over the stinging bee Centaurea hyalolepis and Citrus honeys is noticed which endows the usage of Trigona honey in the antimicrobial industry.
Collapse
Affiliation(s)
- Walid Salem Aburayyan
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Nesrin Seder
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - O’la Al-fawares
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - AbdulFattah Fararjeh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ibrahim S. Majali
- Department of Medical Laboratory Sciences, Mutah University, Karak, Jordan
| | - Yousef Al-Hajaya
- Department of Biological Sciences, Mutah University, Karak, Jordan
| |
Collapse
|
148
|
Trotsko N. Thiazolidin-4-Ones as a Promising Scaffold in the Development of Antibiofilm Agents-A Review. Int J Mol Sci 2023; 25:325. [PMID: 38203498 PMCID: PMC10778874 DOI: 10.3390/ijms25010325] [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: 11/26/2023] [Revised: 12/12/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Thiazolidin-4-ones have a broad range of medical and clinical implementation, which is important for pharmaceutical and medicinal chemistry. This heterocyclic core has been reported to possess a diversity of bioactivities, including antimicrobial and antibiofilm-forming potential. The resistance of biofilms to antibiotics or disinfectants is a serious medical problem. Therefore, there is a natural need to discover new effective structures with properties that inhibit biofilm formation. This review aims to analyze the antibiofilm features of thiazolidin-4-ones described in the literature over the last two decades. The information gathered in this review could benefit the rational design of new effective antibiofilm small molecules with thiazolidin-4-one cores.
Collapse
Affiliation(s)
- Nazar Trotsko
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland
| |
Collapse
|
149
|
Sarkar S, Moitra P, Bhattacharya S. Structure-activity relationship of drug conjugated polymeric materials against uropathogenic bacteria colonization under in vitro and in vivo settings. J Mater Chem B 2023; 12:187-201. [PMID: 38059523 DOI: 10.1039/d3tb01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
The human world has been plagued with different kinds of bacterial infections from time immemorial. The increased development of resistance towards commercial antibiotics has made these bacterial infections an even more critical challenge. Bacteria have modified their mode of interactions with different types of commercial drugs by bringing changes to the receptor proteins or by other resisting mechanisms like drug efflux. Various chemical approaches have been made to date to fight against these smart adapting species. Towards this, we hypothesize chemically modifying the commercial antibacterial drugs in order to deceive the bacteria and destroy the bacterial biomass. In this study, different molecular weight polyethyleneimines are taken and conjugated with some well-known commercial drugs like penicillin and chloramphenicol to explore their antibacterial properties against some of the laboratory and uro-pathogenic strains of Gram-positive and Gram-negative bacteria. A detailed structure-activity relationship of these polymeric prodrug-like materials has been evaluated to determine the optimum formulation. The standardized system not only shows significant ∼90% bacterial killing in liquid broth culture, but also demonstrates promising bacterial inhibition towards biofilm formation for the pathogenic strains on inanimate surfaces like urinary catheters and on an in vivo mouse skin abrasion model. The reported bioactive polymeric materials can be successfully used for widespread therapeutic applications with promising medical relevance.
Collapse
Affiliation(s)
- Sourav Sarkar
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India.
| | - Parikshit Moitra
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Santanu Bhattacharya
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India.
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Srinivasapuram, Yerpedu Mandal, Tirupati District, Andhra Pradesh 517619, India
| |
Collapse
|
150
|
Romero-Montero A, Melgoza-Ramírez LJ, Ruíz-Aguirre JA, Chávez-Santoscoy A, Magaña JJ, Cortés H, Leyva-Gómez G, Del Prado-Audelo ML. Essential-Oils-Loaded Biopolymeric Nanoparticles as Strategies for Microbial and Biofilm Control: A Current Status. Int J Mol Sci 2023; 25:82. [PMID: 38203252 PMCID: PMC10778842 DOI: 10.3390/ijms25010082] [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: 11/09/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
The emergence of bacterial strains displaying resistance to the currently available antibiotics is a critical global concern. These resilient bacteria can form biofilms that play a pivotal role in the failure of bacterial infection treatments as antibiotics struggle to penetrate all biofilm regions. Consequently, eradicating bacteria residing within biofilms becomes considerably more challenging than their planktonic counterparts, leading to persistent and chronic infections. Among various approaches explored, essential oils loaded in nanoparticles based on biopolymers have emerged, promising strategies that enhance bioavailability and biological activities, minimize side effects, and control release through regulated pharmacokinetics. Different available reviews analyze nanosystems and essential oils; however, usually, their main goal is the analysis of their antimicrobial properties, and progress in biofilm combat is rarely discussed, or it is not the primary objective. This review aims to provide a global vision of biofilm conformation and describes mechanisms of action attributed to each EO. Furthermore, we present a comprehensive overview of the latest developments in biopolymeric nanoparticles research, especially in chitosan- and zein-based nanosystems, targeting multidrug-resistant bacteria in both their sessile and biofilm forms, which will help to design precise strategies for combating biofilms.
Collapse
Affiliation(s)
- Alejandra Romero-Montero
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.R.-M.); (G.L.-G.)
| | - Luis Javier Melgoza-Ramírez
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Mexico City 14380, Mexico; (L.J.M.-R.); (J.A.R.-A.); (J.J.M.)
| | - Jesús Augusto Ruíz-Aguirre
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Mexico City 14380, Mexico; (L.J.M.-R.); (J.A.R.-A.); (J.J.M.)
| | - Alejandra Chávez-Santoscoy
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Ave. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico;
| | - Jonathan Javier Magaña
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Mexico City 14380, Mexico; (L.J.M.-R.); (J.A.R.-A.); (J.J.M.)
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.R.-M.); (G.L.-G.)
| | - María Luisa Del Prado-Audelo
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Mexico City 14380, Mexico; (L.J.M.-R.); (J.A.R.-A.); (J.J.M.)
| |
Collapse
|