1
|
Li R, Zhu X, Zhang P, Wu X, Jin Q, Pan J. Ser/Thr protein kinase Stk1 phosphorylates the key transcriptional regulator AlgR to modulate virulence and resistance in Pseudomonas aeruginosa. Virulence 2024; 15:2367649. [PMID: 38898809 PMCID: PMC11197903 DOI: 10.1080/21505594.2024.2367649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
Pseudomonas aeruginosa is one of the leading causes of nosocomial infections worldwide and has emerged as a serious public health threat, due in large part to its multiple virulence factors and remarkable resistance capabilities. Stk1, a eukaryotic-type Ser/Thr protein kinase, has been shown in our previous work to be involved in the regulation of several signalling pathways and biological processes. Here, we demonstrate that deletion of stk1 leads to alterations in several virulence- and resistance-related physiological functions, including reduced pyocyanin and pyoverdine production, attenuated twitching motility, and enhanced biofilm production, extracellular polysaccharide secretion, and antibiotic resistance. Moreover, we identified AlgR, an important transcriptional regulator, as a substrate for Stk1, with its phosphorylation at the Ser143 site catalysed by Stk1. Intriguingly, both the deletion of stk1 and the mutation of Ser143 of AlgR to Ala result in similar changes in the above-mentioned physiological functions. Furthermore, assays of algR expression in these strains suggest that changes in the phosphorylation state of AlgR, rather than its expression level, underlie changes in these physiological functions. These findings uncover Stk1-mediated phosphorylation of AlgR as an important mechanism for regulating virulence and resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Rui Li
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Zhu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Pengfei Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qian Jin
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jianyi Pan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
2
|
van Hoogstraten SWG, Kuik C, Arts JJC, Cillero-Pastor B. Molecular imaging of bacterial biofilms-a systematic review. Crit Rev Microbiol 2024; 50:971-992. [PMID: 37452571 PMCID: PMC11523921 DOI: 10.1080/1040841x.2023.2223704] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
The formation of bacterial biofilms in the human body and on medical devices is a serious human health concern. Infections related to bacterial biofilms are often chronic and difficult to treat. Detailed information on biofilm formation and composition over time is essential for a fundamental understanding of the underlying mechanisms of biofilm formation and its response to anti-biofilm therapy. However, information on the chemical composition, structural components of biofilms, and molecular interactions regarding metabolism- and communication pathways within the biofilm, such as uptake of administered drugs or inter-bacteria communication, remains elusive. Imaging these molecules and their distribution in the biofilm increases insight into biofilm development, growth, and response to environmental factors or drugs. This systematic review provides an overview of molecular imaging techniques used for bacterial biofilm imaging. The techniques included mass spectrometry-based techniques, fluorescence-labelling techniques, spectroscopic techniques, nuclear magnetic resonance spectroscopy (NMR), micro-computed tomography (µCT), and several multimodal approaches. Many molecules were imaged, such as proteins, lipids, metabolites, and quorum-sensing (QS) molecules, which are crucial in intercellular communication pathways. Advantages and disadvantages of each technique, including multimodal approaches, to study molecular processes in bacterial biofilms are discussed, and recommendations on which technique best suits specific research aims are provided.
Collapse
Affiliation(s)
- S. W. G. van Hoogstraten
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, CAPHRI, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - C. Kuik
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht, the Netherlands
| | - J. J. C. Arts
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, CAPHRI, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Biomedical Engineering, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - B. Cillero-Pastor
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht, the Netherlands
- Department of Cell Biology-Inspired Tissue Engineering, The MERLN Institute for Technology-Inspired Regenerative Medicine, University of Maastricht, Maastricht, the Netherlands
| |
Collapse
|
3
|
Lalitha MM, Banerjee S, Jayaraj A, Kamath A, Divakaran D, Yadav V, Lakavathu M, Sajimon J, Anil P, Shaijumon MM, Singh NS, Kurapati R. Two-Dimensional Materials/Biopolymer-Based Antimicrobial Coatings to Thwart Biofilm Formation on Medical Implants. ACS APPLIED BIO MATERIALS 2024; 7:6332-6342. [PMID: 39305253 DOI: 10.1021/acsabm.4c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Infections associated with medical implants due to bacterial adhesion and biofilm formation are a serious problem, leading to acute health risks to patients by compromising their immune system. Therefore, suppressing biofilm formation on biomedical implants is a challenging task, especially for overcoming the drug resistance of bacterial biofilms. Herein, a synergistic efficient surface coating method was developed to inhibit biofilm formation on a model medical implant by combining the antimicrobial property of trimethyl chitosan (TMC) with either 2D material graphene oxide (GO) or black phosphorus (BP) sheets using layer-by-layer (LbL) self-assembly. The multilayer coatings of TMC/GO and TMC/BP were optimized on the glass surface (a model implant) and characterized by using spectroscopic and microscopy techniques. Next, we investigated the antibiofilm formation properties of the TMC/GO and TMC/BP coatings on glass surfaces against both Gram-negative, Escherichia coli (E. coli), and Gram-positive, Bacillus subtilis (B. subtilis), bacteria. The antibiofilm formation was studied using crystal violet (CV) and live/dead assays. Both the live/dead and the CV assays confirmed that the TMC/2D material (2DM)-coated surfaces prevented biofilm formation much more effectively compared to the uncoated surfaces. Scanning electron microscopy analyses revealed that the bacteria were affected physically by incubating with TMC/2DM-coated surfaces due to membrane perturbation, thereby preventing cell attachment and biofilm formation. Further, BP composite coatings (TMC/BP) showed a much better ability to thwart biofilm formation than GO composite coatings (TMC/GO). Also, multilayer coatings showed superior cytocompatibility with human foreskin fibroblast (HFF). Our results demonstrate that the developed coatings TMC/2DMs could be potential candidates for thwarting biofilm formation on medical implants.
Collapse
Affiliation(s)
- Mahesh M Lalitha
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Sourav Banerjee
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Arya Jayaraj
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Adithi Kamath
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Deepika Divakaran
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Vipin Yadav
- School of Physics, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Manikrishna Lakavathu
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Jyothilakshmi Sajimon
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Parvathy Anil
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Manikoth M Shaijumon
- School of Physics, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - N Sadananda Singh
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura 695551, Kerala, India
| |
Collapse
|
4
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
5
|
Hindieh P, Yaghi J, Assaf JC, Chokr A, Atoui A, Louka N, Khoury AE. Unlocking the potential of lactic acid bacteria mature biofilm extracts as antibiofilm agents. AMB Express 2024; 14:112. [PMID: 39361085 PMCID: PMC11450114 DOI: 10.1186/s13568-024-01770-9] [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: 03/05/2024] [Accepted: 09/18/2024] [Indexed: 10/06/2024] Open
Abstract
The continuous growth of biofilm infections and their resilience to conventional cleaning methods and antimicrobial agents pose a worldwide challenge across diverse sectors. This persistent medical, industrial, and environmental issue contributes to treatment challenges and chronic diseases. Lactic acid bacteria have garnered global attention for their substantial antimicrobial effects against pathogens and established beneficial roles. Notably, their biofilms are also predicted to show a promising control strategy against pathogenic biofilm formation. The prevalence of biofilm-related problems underscores the need for extensive research and innovative solutions to tackle this global challenge. This novel study investigates the effect of different extracts (external, internal, and mixed extracts) obtained from Lactobacillus rhamnosus GG biofilm on pathogenic-formed biofilms. Subsequently, external extracts presented an important eradication effectiveness. Furthermore, a 6-fold concentration of these extracts led to eradication percentages of 57%, 67%, and 76% for Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa biofilms, respectively, and around 99.9% bactericidal effect of biofilm cells was observed for the three strains. The results of this research could mark a significant breakthrough in the field of anti-biofilm and antimicrobial strategies. Further studies and molecular research will be necessary to detect the molecules secreted by the biofilm, and their mechanisms of action engaged in new anti-biofilm strategies.
Collapse
Affiliation(s)
- Pamela Hindieh
- Centre d'Analyses et de Recherche (CAR), Unité de Recherche TVA, Laboratoire de mycologie et sécurité des aliments (LMSA), Faculté des Sciences, Université Saint-Joseph de Beyrouth, Campus des sciences et technologies, Mar Roukos, Matn, Lebanon
- Ecole Doctorale "Sciences et Santé", Université Saint-Joseph de Beyrouth, Campus des Sciences Médicales et Infirmières, Riad El Solh, Beirut, Lebanon
| | - Joseph Yaghi
- Centre d'Analyses et de Recherche (CAR), Unité de Recherche TVA, Laboratoire de mycologie et sécurité des aliments (LMSA), Faculté des Sciences, Université Saint-Joseph de Beyrouth, Campus des sciences et technologies, Mar Roukos, Matn, Lebanon
| | - Jean Claude Assaf
- Department of Chemical Engineering, Faculty of Engineering, University of Balamand, P.O. Box 100, Tripoli, 1300, Lebanon.
| | - Ali Chokr
- Research Laboratory of Microbiology (RLM), Department of Life and Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat Campus, Beirut, Lebanon
- Platform of Research and Analysis in Environmental Sciences (PRASE), Doctoral School of Sciences and Technologies, Lebanese University, Hadat Campus, Beirut, Lebanon
| | - Ali Atoui
- Research Laboratory of Microbiology (RLM), Department of Life and Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat Campus, Beirut, Lebanon
| | - Nicolas Louka
- Centre d'Analyses et de Recherche (CAR), Unité de Recherche TVA, Laboratoire de mycologie et sécurité des aliments (LMSA), Faculté des Sciences, Université Saint-Joseph de Beyrouth, Campus des sciences et technologies, Mar Roukos, Matn, Lebanon
| | - André El Khoury
- Centre d'Analyses et de Recherche (CAR), Unité de Recherche TVA, Laboratoire de mycologie et sécurité des aliments (LMSA), Faculté des Sciences, Université Saint-Joseph de Beyrouth, Campus des sciences et technologies, Mar Roukos, Matn, Lebanon
| |
Collapse
|
6
|
Benyamini P. Beyond Antibiotics: What the Future Holds. Antibiotics (Basel) 2024; 13:919. [PMID: 39452186 PMCID: PMC11504868 DOI: 10.3390/antibiotics13100919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
The prevalence of multidrug resistance (MDR) and stagnant drug-development pipelines have led to the rapid rise of hard-to-treat antibiotic-resistant bacterial infections. These infectious diseases are no longer just nosocomial but are also becoming community-acquired. The spread of MDR has reached a crisis level that needs immediate attention. The landmark O'Neill report projects that by 2050, mortality rates associated with MDR bacterial infections will surpass mortality rates associated with individuals afflicted with cancer. Since conventional antimicrobials are no longer very reliable, it is of great importance to investigate different strategies to combat these life-threatening infectious diseases. Here, we provide an overview of recent advances in viable alternative treatment strategies mainly targeting a pathogen's virulence capability rather than viability. Topics include small molecule and immune inhibition of virulence factors, quorum sensing (QS) quenching, inhibition of biofilm development, bacteriophage-mediated therapy, and manipulation of an individual's macroflora to combat MDR bacterial infections.
Collapse
Affiliation(s)
- Payam Benyamini
- Department of Health Sciences at Extension, University of California Los Angeles, 1145 Gayley Ave., Los Angeles, CA 90024, USA
| |
Collapse
|
7
|
Bai LY, Wang ZJ, Lu QY, Huang H, Zhu YY, Zhao YL, Luo XD. 6-Methoxyldihydrochelerythrine Chloride Inhibiting Intra and Extracellular Drug-Resistant Bacteria. ACS Infect Dis 2024; 10:3430-3439. [PMID: 39185798 DOI: 10.1021/acsinfecdis.4c00571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Vancomycin-resistant enterococcus (VRE) is a major nosocomial pathogen that exhibits enhanced infectivity due to its robust virulence and biofilm-forming capabilities. In this study, 6-methoxyldihydrochelerythrine chloride (6-MDC) inhibited the growth of exponential-phase VRE and restored VRE's sensitivity to vancomycin. 6-MDC predominantly suppressed the de novo biosynthetic pathway of pyrimidine and purine in VRE by the RNA-Seq analysis, resulting in obstructed DNA synthesis, which subsequently weakened bacterial virulence and impeded intracellular survival. Furthermore, 6-MDC inhibited biofilm formation, eradicated established biofilms, reduced virulence, and enhanced the host immune response to prevent intracellular survival and replication of VRE. Finally, 6-MDC reduced the VRE load in peritoneal fluid and cells significantly in a murine peritoneal infection model. This paper provides insight into the potential antimicrobial target of benzophenanthridine alkaloids for the first time.
Collapse
Affiliation(s)
- Li-Yu Bai
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Qing-Yu Lu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Huan Huang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, People's Republic of China
| |
Collapse
|
8
|
Richardson JD, Van Lehn RC. Free Energy Analysis of Peptide-Induced Pore Formation in Lipid Membranes by Bridging Atomistic and Coarse-Grained Simulations. J Phys Chem B 2024; 128:8737-8752. [PMID: 39207202 DOI: 10.1021/acs.jpcb.4c03276] [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: 09/04/2024]
Abstract
Antimicrobial peptides (AMPs) are attractive materials for combating the antimicrobial resistance crisis because they can kill target microbes by directly disrupting cell membranes. Although thousands of AMPs have been discovered, their molecular mechanisms of action are still poorly understood. One broad mechanism for membrane disruption is the formation of membrane-spanning hydrophilic pores which can be stabilized by AMPs. In this study, we use molecular dynamics simulations to investigate the thermodynamics of pore formation in model single-component lipid membranes in the presence of one of three AMPs: aurein 1.2, melittin and magainin 2. To overcome the general challenge of modeling long time scale membrane-related behaviors, including AMP binding, clustering, and pore formation, we develop a generalizable methodology for sampling AMP-induced pore formation. This approach involves the long equilibration of peptides around a pore created with a nucleation collective variable by performing coarse-grained simulations, then backmapping equilibrated AMP-membrane configurations to all-atom resolution. We then perform all-atom simulations to resolve free energy profiles for pore formation while accurately modeling the interplay of lipid-peptide-solvent interactions that dictate pore formation free energies. Using this approach, we quantify free energy barriers for pore formation without direct biases on peptides or whole lipids, allowing us to investigate mechanisms of pore formation for these 3 AMPs that are a consequence of unbiased peptide diffusion and clustering. Further analysis of simulation trajectories then relates variations in pore lining by AMPs, AMP-induced lipid disruptions, and salt bridges between AMPs to the observed pore formation free energies and corresponding mechanisms. This methodology and mechanistic analysis have the potential to generalize beyond the AMPs in this study to improve our understanding of pore formation by AMPs and related antimicrobial materials.
Collapse
Affiliation(s)
- Joshua D Richardson
- Department of Chemical and Biological Engineering, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| | - Reid C Van Lehn
- Department of Chemical and Biological Engineering, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
9
|
Jandl B, Dighe S, Gasche C, Makristathis A, Muttenthaler M. Intestinal biofilms: pathophysiological relevance, host defense, and therapeutic opportunities. Clin Microbiol Rev 2024; 37:e0013323. [PMID: 38995034 PMCID: PMC11391705 DOI: 10.1128/cmr.00133-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
SUMMARYThe human intestinal tract harbors a profound variety of microorganisms that live in symbiosis with the host and each other. It is a complex and highly dynamic environment whose homeostasis directly relates to human health. Dysbiosis of the gut microbiota and polymicrobial biofilms have been associated with gastrointestinal diseases, including irritable bowel syndrome, inflammatory bowel diseases, and colorectal cancers. This review covers the molecular composition and organization of intestinal biofilms, mechanistic aspects of biofilm signaling networks for bacterial communication and behavior, and synergistic effects in polymicrobial biofilms. It further describes the clinical relevance and diseases associated with gut biofilms, the role of biofilms in antimicrobial resistance, and the intestinal host defense system and therapeutic strategies counteracting biofilms. Taken together, this review summarizes the latest knowledge and research on intestinal biofilms and their role in gut disorders and provides directions toward the development of biofilm-specific treatments.
Collapse
Affiliation(s)
- Bernhard Jandl
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Satish Dighe
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christoph Gasche
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- Loha for Life, Center for Gastroenterology and Iron Deficiency, Vienna, Austria
| | - Athanasios Makristathis
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
10
|
Lee H, Hwang SH, Shin H, Ha NC, Wang Q, Choi SH. Identification and characterization of a small molecule BFstatin inhibiting BrpR, the transcriptional regulator for biofilm formation of Vibrio vulnificus. Front Microbiol 2024; 15:1468567. [PMID: 39314881 PMCID: PMC11416940 DOI: 10.3389/fmicb.2024.1468567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Many pathogenic bacteria form biofilms that are resistant to not only host immune defenses but also antibiotics, posing a need for the development of strategies to control biofilms. In this study, to prevent biofilm formation of the fulminating foodborne pathogen Vibrio vulnificus, chemical libraries were extensively screened to identify a small molecule inhibiting the activity of BrpR, a transcriptional regulator for biofilm genes. Accordingly, the BrpR inhibitor BFstatin [N1-(2-chloro-5-fluorophenyl)-N3-propylmalonamide], with a half-maximal effective concentration of 8.01 μM, was identified. BFstatin did not interfere with bacterial growth or exhibit cytotoxicity to the human epithelial cell line. BFstatin directly bound to BrpR and interrupted its binding to the target promoter DNAs of the downstream genes. Molecular dynamics simulation of the interaction between BFstatin and BrpR proposed that BFstatin modifies the structure of BrpR, especially the DNA-binding domain. Transcriptomic analyses revealed that BFstatin reduces the expression of the BrpR regulon including the cabABC operon and brp locus which contribute to the production of biofilm matrix of V. vulnificus. Accordingly, BFstatin diminished the biofilm levels of V. vulnificus by inhibiting the matrix development in a concentration-dependent manner. Altogether, BFstatin could be an anti-biofilm agent targeting BrpR, thereby rendering V. vulnificus more susceptible to host immune defenses and antibiotics.
Collapse
Affiliation(s)
- Hojun Lee
- Department of Agricultural Biotechnology, National Research Laboratory of Molecular Microbiology and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Seung-Ho Hwang
- Department of Agricultural Biotechnology, National Research Laboratory of Molecular Microbiology and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Hyunwoo Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Nam-Chul Ha
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Sang Ho Choi
- Department of Agricultural Biotechnology, National Research Laboratory of Molecular Microbiology and Toxicology, Seoul National University, Seoul, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Cusumano JA, Kalogeropoulos AP, Le Provost M, Gallo NR, Levine SM, Inzana T, Papamanoli A. The emerging challenge of Enterococcus faecalis endocarditis after transcatheter aortic valve implantation: time for innovative treatment approaches. Clin Microbiol Rev 2024:e0016823. [PMID: 39235238 DOI: 10.1128/cmr.00168-23] [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: 09/06/2024] Open
Abstract
SUMMARYInfective endocarditis (IE) is a life-threatening infection that has nearly doubled in prevalence over the last two decades due to the increase in implantable cardiac devices. Transcatheter aortic valve implantation (TAVI) is currently one of the most common cardiac procedures. TAVI usage continues to exponentially rise, inevitability increasing TAVI-IE. Patients with TAVI are frequently nonsurgical candidates, and TAVI-IE 1-year mortality rates can be as high as 74% without valve or bacterial biofilm removal. Enterococcus faecalis, a historically less common IE pathogen, is the primary cause of TAVI-IE. Treatment options are limited due to enterococcal intrinsic resistance and biofilm formation. Novel approaches are warranted to tackle current therapeutic gaps. We describe the existing challenges in treating TAVI-IE and how available treatment discovery approaches can be combined with an in silico "Living Heart" model to create solutions for the future.
Collapse
Affiliation(s)
- Jaclyn A Cusumano
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York, USA
| | - Andreas P Kalogeropoulos
- Renaissance School of Medicine Division of Cardiology, Stony Brook University, Stony Brook, New York, USA
| | - Mathieu Le Provost
- School of Engineering, Computer Science and Artificial Intelligence, Long Island University, Brooklyn, New York, USA
| | - Nicolas R Gallo
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York, USA
- School of Engineering, Computer Science and Artificial Intelligence, Long Island University, Brooklyn, New York, USA
| | | | - Thomas Inzana
- College of Veterinary Medicine, Long Island University, Brooklyn, New York, USA
| | - Aikaterini Papamanoli
- Division of Infectious Diseases, Stony Brook University Medical Center, Stony Brook, New York, USA
| |
Collapse
|
12
|
Tanwar SN, Parauha YR, There Y, Ameen F, Dhoble SJ. Inorganic nanoparticles: An effective antibiofilm strategy. LUMINESCENCE 2024; 39:e4878. [PMID: 39223925 DOI: 10.1002/bio.4878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/08/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Biofilm is a common problem associated with human health. Pathogenicity and increase in resistance of bacteria require urgent development of effective ways for the treatment of bacterial diseases. Different strategies have been developed for the treatment of bacterial infections among which nanoparticles have shown greater prospects in battling with infections. Biofilms are resistant microbial colonies that possess resistance and, hence, cannot be killed by conventional drugs. Nanoparticles offer new avenues for treating biofilm-related infections involving multi-drug resistant organisms. They possess great antibiofilm properties, disrupting cell architecture and preventing colony formation. Green-synthesised nanoparticles are more effective and less toxic to human cells than commercially available or chemically synthesised antibiofilm nanoparticles. This review summarises the antibiofilm efficiency of plant-mediated nanoparticles and knowledge about biofilm inhibition.
Collapse
Affiliation(s)
- Shruti Nandkishor Tanwar
- Department of Microbiology, Taywade College, Mahadula-Koradi, Nagpur, India
- Department of Physics, R.T.M., Nagpur University, Nagpur, India
| | - Yatish Ratn Parauha
- Department of Physics, Shri Ramdeobaba College of Engineering and Management, Nagpur, India
- Ramdeobaba University, Nagpur, India
| | - Yogesh There
- Department of Microbiology, Taywade College, Mahadula-Koradi, Nagpur, India
| | - Faud Ameen
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh, Saudi Arbia
| | | |
Collapse
|
13
|
Repac Antić D, Kovač B, Kolenc M, Brčić Karačonji I, Gobin I, Petković Didović M. Combinatory Effect of Nitroxoline and Gentamicin in the Control of Uropathogenic Enterococci Infections. Antibiotics (Basel) 2024; 13:829. [PMID: 39335003 PMCID: PMC11428728 DOI: 10.3390/antibiotics13090829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Enterococcus faecalis, responsible for a majority of human and nosocomial enterococcal infections, is intrinsically resistant to aminoglycoside antibiotics (such as gentamicin, GEN), which must be used in a combined therapy to be effective. Nitroxoline (NTX) is an old antibiotic, underused for decades, but rediscovered now in an era of growing antibiotic resistance. In this in vitro study, the types of interactions between NTX and GEN on 29 E. faecalis strains were analyzed with an aim to find synergistic antimicrobial and antiadhesion combinations. Transmission electron microscopy (TEM) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) were used to analyze changes in cell morphology and bacterial proteome after monotreatments and combined treatments. The results showed the synergistic effect for six combinations on eight strains, including the ATCC29212, and an additive effect for most strains. Combinations causing a complete inhibition of adhesion were established. Cell membrane integrity was affected by NTX, while combined NTX/GEN treatment caused dramatic changes in cell morphology. Upregulation of the expression of many proteins was established, with some emerging only after combined treatment. The results strongly imply that NTX has the potential for use in combined therapy with GEN against enterococci and it could further provide a substantial contribution to an ongoing fight against antimicrobial resistance and nosocomial infections.
Collapse
Affiliation(s)
- Davorka Repac Antić
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Department of Clinical Microbiology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Bruno Kovač
- Chair of Buildings and Constructional Complexes, Faculty of Civil and Geodetic Engineering, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Marko Kolenc
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Irena Brčić Karačonji
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
- Department of Basic Medical Sciences, Faculty of Health Studies, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Gobin
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Mirna Petković Didović
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
14
|
De Padua JC, Tanaka T, Ueno K, Dela Cruz TEE, Ishihara A. Isolation of 2,2'-azoxybisbenzyl alcohol from Agaricus subrutilescens and its inhibitory activity against bacterial biofilm formation. Biosci Biotechnol Biochem 2024; 88:983-991. [PMID: 38925646 DOI: 10.1093/bbb/zbae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Virulence pathways in pathogenic bacteria are regulated by quorum sensing mechanisms, particularly biofilm formation through autoinducer (AI) production and sensing. In this study, the culture filtrate extracted from an edible mushroom, Agaricus subrutilescens, was fractionated to isolate a compound that inhibits biofilm formation. Four gram-negative bacteria (Klebsiella pneumoniae, Escherichia coli, Proteus mirabilis, and Enterobacter cloacae) and two gram-positive bacteria (Enterococcus faecalis and Staphylococcus aureus) were used for the bioassay. The bioassay-guided chromatographic separations of the culture filtrate extract resulted in the isolation of the compound. Further, spectroscopic analyses revealed the identity of the compound as 2,2'-azoxybisbenzyl alcohol (ABA). The minimum inhibitory and sub-inhibitory concentrations of the compound were also determined. Azoxybisbenzyl alcohol was significantly effective in inhibiting biofilm formation in all tested bacteria, with half-maximal inhibitory concentrations of 3-11 µg/mL. Additionally, the bioactivity of ABA was confirmed through the bioassays for the inhibition of exopolysaccharide matrixes and AI activities.
Collapse
Affiliation(s)
- Jewel C De Padua
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, Japan
| | - Tomoya Tanaka
- Graduate School of Sustainability Sciences, Tottori University, Tottori, Japan
| | - Kotomi Ueno
- Department of Agricultural, Life, Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Thomas Edison E Dela Cruz
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
| | - Atsushi Ishihara
- Department of Agricultural, Life, Environmental Sciences, Faculty of Agriculture, Tottori University, Tottori, Japan
- Fungus/Mushroom Resource and Research Center, Faculty of Agriculture, Tottori, Japan
| |
Collapse
|
15
|
Lee JH, Kim YG, Choi JS, Jeong YT, Hwang BS, Lee J. Antibiofilm and Antihemolytic Activities of Actinostemma lobatum Extract Rich in Quercetin against Staphylococcus aureus. Pharmaceutics 2024; 16:1075. [PMID: 39204420 PMCID: PMC11359957 DOI: 10.3390/pharmaceutics16081075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Staphylococcus aureus biofilm formation is a pivotal mechanism in the development of drug resistance, conferring resilience against conventional antibiotics. This study investigates the inhibitory effects of Actinostemma lobatum (A. lobatum) Maxim extracts on S. aureus biofilm formation and their antihemolytic activities, with a particular focus on identifying the active antibiofilm and antihemolysis compound, quercetin. Seven solvent extracts and twelve sub-fractions were evaluated against four S. aureus strains. The ethyl acetate fraction (10 to 100 μg/mL) significantly hindered biofilm formation by both methicillin-sensitive and -resistant strains. Bioassay-guided isolation of the ethyl acetate extract identified quercetin as the major antibiofilm compound. The ethyl acetate extract was found to contain 391 μg/mg of quercetin and 30 μg/mg of kaempferol. Additionally, the A. lobatum extract exhibited antihemolytic activity attributable to the presence of quercetin. The findings suggest that quercetin-rich extracts from A. lobatum and other quercetin-rich foods and plants hold promise for inhibiting resilient S. aureus biofilm formation and attenuating its virulence.
Collapse
Affiliation(s)
- Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (J.-H.L.); (Y.-G.K.)
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (J.-H.L.); (Y.-G.K.)
| | - Ji-Su Choi
- Nakdonggang National Institute of Biological Resources, 137, Donam 2-gil, Sangju-si 37242, Republic of Korea; (J.-S.C.); (Y.T.J.)
| | - Yong Tae Jeong
- Nakdonggang National Institute of Biological Resources, 137, Donam 2-gil, Sangju-si 37242, Republic of Korea; (J.-S.C.); (Y.T.J.)
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, 137, Donam 2-gil, Sangju-si 37242, Republic of Korea; (J.-S.C.); (Y.T.J.)
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (J.-H.L.); (Y.-G.K.)
| |
Collapse
|
16
|
Admane N, Kothandan R, Biswas S. Amyloid transformations of phenol soluble modulin α1 in Staphylococcus aureus and their modulation deploying a prenylated chalcone. Sci Rep 2024; 14:18587. [PMID: 39127763 PMCID: PMC11316821 DOI: 10.1038/s41598-024-69344-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024] Open
Abstract
Phenol soluble modulins (PSMs) are small amphipathic peptides involved in a series of biological functions governing staphylococcal pathogenesis, primarily by facilitating the formation of an extracellular fibril structure with amyloid-like properties. This fibrillar architecture stabilizes the staphylococcal biofilm making it resilient to antibiotic treatment. Our study aims to abrogate the amyloid fibrillation of PSM α1 with novel insights on the amyloid modulatory potential of a prenylated chalcone, Isobavachalcone (IBC). A combination of biophysical and computational assays to address the amyloid modulatory effect of IBC has been undertaken to arrive at a model for the inhibition of PSM α1 fibrillation. ThT kinetics studies indicated that IBC must be stably interacting with the amyloidogenic core of PSM α1 monomers or it may be inhibiting the pre-fibrillar aggregates populated at the early stages of amyloid transformation kinetics. This heteromolecular association further inhibits the amyloid transformation corroborated by a ∼ 94% and ∼ 91% reduction in the ThT maxima, even at sub-stoichiometric concentrations. Transmission electron microscopy (TEM) of end-stage aggregates (∼ 55 h) depict mature, inter-twined, laterally stacked amyloid fibrils in untreated PSM α1 samples while this fibrillar load is remarkably reduced in the presence of IBC. The inhibitory effect of IBC on the β-sheet transitions of PSM α1 were also validated using far-UV CD spectra. Molecular dynamics simulation studies with PSM aggregates (PSM-A) have also suggested that IBC disrupts the hydrogen bonding interactions and corroborates the inhibition of alpha to beta transitions of PSM-A. Collectively, our data proposes a novel structural motif for the rational discovery of non-toxic therapeutic agents targeting the functional amyloids which have slowly emerged as potent factors, consolidating the antibiotic resistant staphylococcal biofilm assembly.
Collapse
Affiliation(s)
- Nikita Admane
- ViStA Laboratory, Department of Biological Sciences, BITS, Pilani - KK Birla Goa Campus, Goa, 403726, India
| | - Ram Kothandan
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, India
| | - Sumit Biswas
- ViStA Laboratory, Department of Biological Sciences, BITS, Pilani - KK Birla Goa Campus, Goa, 403726, India.
| |
Collapse
|
17
|
Mini M, Jayakumar D, Kumar P. In-silico and in-vitro assessment of the antibiofilm potential of azo dye, carmoisine against Pseudomonas aeruginosa. J Biomol Struct Dyn 2024; 42:6700-6710. [PMID: 37485898 DOI: 10.1080/07391102.2023.2237579] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
Biofilm is a community of microorganisms attached to the substrate and plays a significant role in microbial pathogenesis and medical device-related infection. Pseudomonas aeruginosa (PA) is a highly infectious gram-negative opportunistic biofilm-forming bacterium with high antibiotic resistance. Several reports underscore the antimicrobial activity of natural and synthetic food coloring agents, including carmoisine, turmeric dye, red amaranth dye, and phloxine B. However, their ability to suppress the PA biofilm is not clearly understood. Carmoisine is a red-colored synthetic azo dye containing naphthalene subunits and sulfonic groups and is widely used as a food coloring agent. This study investigated the antibiofilm potential and possible mechanism of biofilm inhibition by carmoisine against PA. Computational studies through molecular docking revealed that carmoisine strongly binds to QS regulator LasR (-12.7) and relatively less strongly but significantly with WspR (-6.9). Further analysis of the docked LasR-carmoisine complex using 100 ns MD simulation (Desmond, Schrödinger) validated the bonding strength and stability. Crystal violet assay, triphenyl tetrazolium chloride salt assay, and confocal microscopic studies were adopted for biofilm quantification, and the results indicated the dose-dependent antibiofilm activity of carmoisine against PA. We hypothesise that the carmoisine-mediated reduction of biofilm in PA is due to its interaction with LasR and interference with the QS system. The computational and biochemical analysis of another compound, 1,2-naphthoquinone-4-sulphonic acid, reiterated the role of the naphthalene ring in biofilm inhibition. Hence, this work will pave the way for the future discovery of antibiofilm drugs based on naphthalene ring-based lead compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Minsa Mini
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Devi Jayakumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Praveen Kumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| |
Collapse
|
18
|
Kozień Ł, Policht A, Heczko P, Arent Z, Bracha U, Pardyak L, Pietsch-Fulbiszewska A, Gallienne E, Piwowar P, Okoń K, Tomusiak-Plebanek A, Strus M. PDIA iminosugar influence on subcutaneous Staphylococcus aureus and Pseudomonas aeruginosa infections in mice. Front Cell Infect Microbiol 2024; 14:1395577. [PMID: 39145303 PMCID: PMC11322076 DOI: 10.3389/fcimb.2024.1395577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction Biofilm-associated infections persist as a therapeutic challenge in contemporary medicine. The efficacy of antibiotic therapies is ineffective in numerous instances, necessitating a heightened focus on exploring novel anti-biofilm medical strategies. Among these, iminosugars emerge as a distinctive class of compounds displaying promising biofilm inhibition properties. Methods This study employs an in vivo wound infection mouse model to evaluate the effectiveness of PDIA in treating biofilm-associated skin wound infections caused by Staphylococcus aureus and Pseudomonas aeruginosa. Dermic wounds in mice were infected with biofilm-forming strains, specifically S. aureus 48 and P. aeruginosa 5, which were isolated from patients with diabetic foot, and are well-known for their strong biofilm formation. The subsequent analysis included clinical, microbiological, and histopathological parameters. Furthermore, an exploration into the susceptibility of the infectious strains to hydrogen peroxide was conducted, acknowledging its potential presence during induced inflammation in mouse dermal wounds within an in vivo model. Results The findings revealed the efficacy of PDIA iminosugar against the S. aureus strain, evidenced by a reduction in bacterial numbers within the wound and the inflammatory focus. Discussion This study suggests that PDIA iminosugar emerges as an active and potentially effective antibiofilm agent, positioning it as a viable treatment option for staphylococcal infections.
Collapse
Affiliation(s)
- Łucja Kozień
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Aleksandra Policht
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Piotr Heczko
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Zbigniew Arent
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Urszula Bracha
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Laura Pardyak
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Agnieszka Pietsch-Fulbiszewska
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Estelle Gallienne
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311, Université d'Orléans & CNRS, Orléans, France
| | - Piotr Piwowar
- Faculty of Electrical Engineering, Automatics, Computer Science and Biomedical Engineering, AGH University of Science and Technology, Kraków, Poland
| | - Krzysztof Okoń
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Tomusiak-Plebanek
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Magdalena Strus
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
19
|
Fu J, Wang D, Tang Z, Xu Y, Xie J, Chen R, Wang P, Zhong Q, Ning Y, Lei M, Mai H, Li H, Liu H, Wang J, Cheng H. NIR-responsive electrospun nanofiber dressing promotes diabetic-infected wound healing with programmed combined temperature-coordinated photothermal therapy. J Nanobiotechnology 2024; 22:384. [PMID: 38951903 PMCID: PMC11218286 DOI: 10.1186/s12951-024-02621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Diabetic wounds present significant challenges, specifically in terms of bacterial infection and delayed healing. Therefore, it is crucial to address local bacterial issues and promote accelerated wound healing. In this investigation, we utilized electrospinning to fabricate microgel/nanofiber membranes encapsulating MXene-encapsulated microgels and chitosan/gelatin polymers. RESULTS The film dressing facilitates programmed photothermal therapy (PPT) and mild photothermal therapy (MPTT) under near-infrared (NIR), showcasing swift and extensive antibacterial and biofilm-disrupting capabilities. The PPT effect achieves prompt sterilization within 5 min at 52 °C and disperses mature biofilm within 10 min. Concurrently, by adjusting the NIR power to induce local mild heating (42 °C), the dressing stimulates fibroblast proliferation and migration, significantly enhancing vascularization. Moreover, in vivo experimentation successfully validates the film dressing, underscoring its immense potential in addressing the intricacies of diabetic wounds. CONCLUSIONS The MXene microgel-loaded nanofiber dressing employs temperature-coordinated photothermal therapy, effectively amalgamating the advantageous features of high-temperature sterilization and low-temperature promotion of wound healing. It exhibits rapid, broad-spectrum antibacterial and biofilm-disrupting capabilities, exceptional biocompatibility, and noteworthy effects on promoting cell proliferation and vascularization. These results affirm the efficacy of our nanofiber dressing, highlighting its significant potential in addressing the challenge of diabetic wounds struggling to heal due to infection.
Collapse
Affiliation(s)
- Jinlang Fu
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ding Wang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zinan Tang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yixin Xu
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiajun Xie
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Rong Chen
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pinkai Wang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, China
| | - Qiang Zhong
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanhong Ning
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mingyuan Lei
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huaming Mai
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Li
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haibing Liu
- Department of Orthopaedic, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, Hunan, 421001, China.
| | - Jian Wang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Hao Cheng
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
20
|
da Cruz Nizer WS, Adams ME, Allison KN, Montgomery MC, Mosher H, Cassol E, Overhage J. Oxidative stress responses in biofilms. Biofilm 2024; 7:100203. [PMID: 38827632 PMCID: PMC11139773 DOI: 10.1016/j.bioflm.2024.100203] [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: 02/28/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/04/2024] Open
Abstract
Oxidizing agents are low-molecular-weight molecules that oxidize other substances by accepting electrons from them. They include reactive oxygen species (ROS), such as superoxide anions (O2-), hydrogen peroxide (H2O2), and hydroxyl radicals (HO-), and reactive chlorine species (RCS) including sodium hypochlorite (NaOCl) and its active ingredient hypochlorous acid (HOCl), and chloramines. Bacteria encounter oxidizing agents in many different environments and from diverse sources. Among them, they can be produced endogenously by aerobic respiration or exogenously by the use of disinfectants and cleaning agents, as well as by the mammalian immune system. Furthermore, human activities like industrial effluent pollution, agricultural runoff, and environmental activities like volcanic eruptions and photosynthesis are also sources of oxidants. Despite their antimicrobial effects, bacteria have developed many mechanisms to resist the damage caused by these toxic molecules. Previous research has demonstrated that growing as a biofilm particularly enhances bacterial survival against oxidizing agents. This review aims to summarize the current knowledge on the resistance mechanisms employed by bacterial biofilms against ROS and RCS, focussing on the most important mechanisms, including the formation of biofilms in response to oxidative stressors, the biofilm matrix as a protective barrier, the importance of detoxifying enzymes, and increased protection within multi-species biofilm communities. Understanding the complexity of bacterial responses against oxidative stress will provide valuable insights for potential therapeutic interventions and biofilm control strategies in diverse bacterial species.
Collapse
Affiliation(s)
| | - Madison Elisabeth Adams
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | - Kira Noelle Allison
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | | | - Hailey Mosher
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| |
Collapse
|
21
|
Kosmeri C, Giapros V, Serbis A, Balomenou F, Baltogianni M. Antibiofilm Strategies in Neonatal and Pediatric Infections. Antibiotics (Basel) 2024; 13:509. [PMID: 38927176 PMCID: PMC11200539 DOI: 10.3390/antibiotics13060509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Biofilm-related infections pose significant challenges in neonatal and pediatric care, contributing to increased morbidity and mortality rates. These complex microbial communities, comprising bacteria and fungi, exhibit resilience against antibiotics and host immune responses. Bacterial species such as Enterococcus faecalis, Pseudomonas aeruginosa, Staphylococcus aureus, and Staphylococcus epidermidis commonly form biofilms on medical devices, exacerbating infection risks. Neonates and children, particularly those in intensive care units, are highly susceptible to biofilm-associated infections due to the prolonged use of invasive devices, such as central lines and endotracheal tubes. Enteral feeding tubes, crucial for neonatal nutritional support, also serve as potential sites for biofilm formation, contributing to recurrent microbial contamination. Moreover, Candida species, including Candida pelliculosa, present emerging challenges in neonatal care, with multi-drug resistant strains posing treatment complexities. Current antimicrobial therapies, while important in managing infections, often fall short in eradicating biofilms, necessitating alternative strategies. The aim of this review is to summarize current knowledge regarding antibiofilm strategies in neonates and in children. Novel approaches focusing on biofilm inhibition and dispersal show promise, including surface modifications, matrix-degrading enzymes, and quorum-sensing inhibitors. Prudent use of medical devices and exploration of innovative antibiofilm therapies are imperative in mitigating neonatal and pediatric biofilm infections.
Collapse
Affiliation(s)
- Chrysoula Kosmeri
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece; (C.K.); (A.S.)
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| | - Anastasios Serbis
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece; (C.K.); (A.S.)
| | - Foteini Balomenou
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| | - Maria Baltogianni
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| |
Collapse
|
22
|
Cho JA, Jeon S, Kwon Y, Roh YJ, Lee CH, Kim SJ. Comparative proteomics analysis of biofilms and planktonic cells of Enterococcus faecalis and Staphylococcus lugdunensis with contrasting biofilm-forming ability. PLoS One 2024; 19:e0298283. [PMID: 38809833 PMCID: PMC11135667 DOI: 10.1371/journal.pone.0298283] [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: 07/26/2023] [Accepted: 01/18/2024] [Indexed: 05/31/2024] Open
Abstract
Biofilms make it difficult to eradicate bacterial infections through antibiotic treatments and lead to numerous complications. Previously, two periprosthetic infection-related pathogens, Enterococcus faecalis and Staphylococcus lugdunensis were reported to have relatively contrasting biofilm-forming abilities. In this study, we examined the proteomics of the two microorganisms' biofilms using LC-MS/MS. The results showed that each microbe exhibited an overall different profile for differential gene expressions between biofilm and planktonic cells as well as between each other. Of a total of 929 proteins identified in the biofilms of E. faecalis, 870 proteins were shared in biofilm and planktonic cells, and 59 proteins were found only in the biofilm. In S. lugdunensis, a total of 1125 proteins were identified, of which 1072 proteins were found in common in the biofilm and planktonic cells, and 53 proteins were present only in the biofilms. The functional analysis for the proteins identified only in the biofilms using UniProt keywords demonstrated that they were mostly assigned to membrane, transmembrane, and transmembrane helix in both microorganisms, while hydrolase and transferase were found only in E. faecalis. Protein-protein interaction analysis using STRING-db indicated that the resulting networks did not have significantly more interactions than expected. GO term analysis exhibited that the highest number of proteins were assigned to cellular process, catalytic activity, and cellular anatomical entity. KEGG pathway analysis revealed that microbial metabolism in diverse environments was notable for both microorganisms. Taken together, proteomics data discovered in this study present a unique set of biofilm-embedded proteins of each microorganism, providing useful information for diagnostic purposes and the establishment of appropriately tailored treatment strategies. Furthermore, this study has significance in discovering the target candidate molecules to control the biofilm-associated infections of E. faecalis and S. lugdunensis.
Collapse
Affiliation(s)
- Jung-Ah Cho
- Department of Orthopedic Surgery, Dongtan Sacred Hospital, Hallym University, Hwaseong, Republic of Korea
- College of Transdisciplinary Studies, School of Undergraduate Studies, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Sangsoo Jeon
- College of Transdisciplinary Studies, School of Undergraduate Studies, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Youngmin Kwon
- Department of Orthopedic Surgery, Dongtan Sacred Hospital, Hallym University, Hwaseong, Republic of Korea
| | - Yoo Jin Roh
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Chang-Hun Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Sung Jae Kim
- Department of Orthopedic Surgery, Dongtan Sacred Hospital, Hallym University, Hwaseong, Republic of Korea
| |
Collapse
|
23
|
Casaro S, Prim JG, Gonzalez TD, Cunha F, Bisinotto RS, Chebel RC, Santos JEP, Nelson CD, Jeon SJ, Bicalho RC, Driver JP, Galvão KN. Integrating uterine microbiome and metabolome to advance the understanding of the uterine environment in dairy cows with metritis. Anim Microbiome 2024; 6:30. [PMID: 38802977 PMCID: PMC11131188 DOI: 10.1186/s42523-024-00314-7] [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: 01/25/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Metritis is a prevalent uterine disease that affects the welfare, fertility, and survival of dairy cows. The uterine microbiome from cows that develop metritis and those that remain healthy do not differ from calving until 2 days postpartum, after which there is a dysbiosis of the uterine microbiome characterized by a shift towards opportunistic pathogens such as Fusobacteriota and Bacteroidota. Whether these opportunistic pathogens proliferate and overtake the uterine commensals could be determined by the type of substrates present in the uterus. The objective of this study was to integrate uterine microbiome and metabolome data to advance the understanding of the uterine environment in dairy cows that develop metritis. Holstein cows (n = 104) had uterine fluid collected at calving and at the day of metritis diagnosis. Cows with metritis (n = 52) were paired with cows without metritis (n = 52) based on days after calving. First, the uterine microbiome and metabolome were evaluated individually, and then integrated using network analyses. RESULTS The uterine microbiome did not differ at calving but differed on the day of metritis diagnosis between cows with and without metritis. The uterine metabolome differed both at calving and on the day of metritis diagnosis between cows that did and did not develop metritis. Omics integration was performed between 6 significant bacteria genera and 153 significant metabolites on the day of metritis diagnosis. Integration was not performed at calving because there were no significant differences in the uterine microbiome. A total of 3 bacteria genera (i.e. Fusobacterium, Porphyromonas, and Bacteroides) were strongly correlated with 49 metabolites on the day of metritis diagnosis. Seven of the significant metabolites at calving were among the 49 metabolites strongly correlated with opportunistic pathogenic bacteria on the day of metritis diagnosis. The main metabolites have been associated with attenuation of biofilm formation by commensal bacteria, opportunistic pathogenic bacteria overgrowth, tissue damage and inflammation, immune evasion, and immune dysregulation. CONCLUSIONS The data integration presented herein helps advance the understanding of the uterine environment in dairy cows with metritis. The identified metabolites may provide a competitive advantage to the main uterine pathogens Fusobacterium, Porphyromonas and Bacteroides, and may be promising targets for future interventions aiming to reduce opportunistic pathogenic bacteria growth in the uterus.
Collapse
Affiliation(s)
- S Casaro
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - J G Prim
- Department of Clinical Sciences, Auburn University, Auburn, AL, USA
| | - T D Gonzalez
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - F Cunha
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - R S Bisinotto
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - R C Chebel
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - J E P Santos
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
- D. H. Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL, USA
| | - C D Nelson
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - S J Jeon
- Department of Veterinary Biomedical Sciences, Long Island University, Brookville, NY, USA
| | - R C Bicalho
- FERA Diagnostics and Biologicals, College Station, TX, USA
| | - J P Driver
- Division of Animals Sciences, University of Missouri, Columbia, MO, USA
| | - Klibs N Galvão
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, USA.
- D. H. Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
24
|
Fik-Jaskółka M, Mittova V, Motsonelidze C, Vakhania M, Vicidomini C, Roviello GN. Antimicrobial Metabolites of Caucasian Medicinal Plants as Alternatives to Antibiotics. Antibiotics (Basel) 2024; 13:487. [PMID: 38927153 PMCID: PMC11200912 DOI: 10.3390/antibiotics13060487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
This review explores the potential of antimicrobial metabolites derived from Caucasian medicinal plants as alternatives to conventional antibiotics. With the rise of antibiotic resistance posing a global health threat, there is a pressing need to investigate alternative sources of antimicrobial agents. Caucasian medicinal plants have traditionally been used for their therapeutic properties, and recent research has highlighted their potential as sources of antimicrobial compounds. Representatives of 15 families of Caucasian medicinal plant extracts (24 species) have been explored for their efficacy against these pathogens. The effect of these plants on Gram-positive and Gram-negative bacteria and fungi is discussed in this paper. By harnessing the bioactive metabolites present in these plants, this study aims to contribute to the development of new antimicrobial treatments that can effectively combat bacterial infections while minimizing the risk of resistance emergence. Herein we discuss the following classes of bioactive compounds exhibiting antimicrobial activity: phenolic compounds, flavonoids, tannins, terpenes, saponins, alkaloids, and sulfur-containing compounds of Allium species. The review discusses the pharmacological properties of selected Caucasian medicinal plants, the extraction and characterization of these antimicrobial metabolites, the mechanisms of action of antibacterial and antifungal plant compounds, and their potential applications in clinical settings. Additionally, challenges and future directions in the research of antimicrobial metabolites from Caucasian medicinal plants are addressed.
Collapse
Affiliation(s)
- Marta Fik-Jaskółka
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Valentina Mittova
- Teaching University Geomedi, 4 King Solomon II Str., Tbilisi 0114, Georgia; (V.M.)
| | | | - Malkhaz Vakhania
- Teaching University Geomedi, 4 King Solomon II Str., Tbilisi 0114, Georgia; (V.M.)
| | - Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| |
Collapse
|
25
|
Han D, Ma S, He C, Yang Y, Li P, Lu L. Unveiling the genetic architecture and transmission dynamics of a novel multidrug-resistant plasmid harboring bla NDM-5 in E. Coli ST167: implications for antibiotic resistance management. BMC Microbiol 2024; 24:178. [PMID: 38783210 PMCID: PMC11112900 DOI: 10.1186/s12866-024-03333-1] [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: 02/26/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The emergence of multidrug-resistant (MDR) Escherichia coli strains poses significant challenges in clinical settings, particularly when these strains harbor New Delhi metallo-ß-lactamase (NDM) gene, which confer resistance to carbapenems, a critical class of last-resort antibiotics. This study investigates the genetic characteristics and implications of a novel blaNDM-5-carrying plasmid pNDM-5-0083 isolated from an E. coli strain GZ04-0083 from clinical specimen in Zhongshan, China. RESULTS Phenotypic and genotypic evaluations confirmed that the E. coli ST167 strain GZ04-0083 is a multidrug-resistant organism, showing resistance to diverse classes of antibiotics including ß-lactams, carbapenems, fluoroquinolones, aminoglycosides, and sulfonamides, while maintaining susceptibility to monobactams. Investigations involving S1 pulsed-field gel electrophoresis, Southern blot analysis, and conjugation experiments, alongside genomic sequencing, confirmed the presence of the blaNDM-5 gene within a 146-kb IncFIB plasmid pNDM-5-0083. This evidence underscores a significant risk for the horizontal transfer of resistance genes among bacterial populations. Detailed annotations of genetic elements-such as resistance genes, transposons, and insertion sequences-and comparative BLAST analyses with other blaNDM-5-carrying plasmids, revealed a unique architectural configuration in the pNDM-5-0083. The MDR region of this plasmid shares a conserved gene arrangement (repA-IS15DIV-blaNDM-5-bleMBL-IS91-suI2-aadA2-dfrA12) with three previously reported plasmids, indicating a potential for dynamic genetic recombination and evolution within the MDR region. Additionally, the integration of virulence factors, including the iro and sit gene clusters and enolase, into its genetic architecture poses further therapeutic challenges by enhancing the strain's pathogenicity through improved host tissue colonization, immune evasion, and increased infection severity. CONCLUSIONS The detailed identification and characterization of pNDM-5-0083 enhance our understanding of the mechanisms facilitating the spread of carbapenem resistance. This study illuminates the intricate interplay among various genetic elements within the novel blaNDM-5-carrying plasmid, which are crucial for the stability and mobility of resistance genes across bacterial populations. These insights highlight the urgent need for ongoing surveillance and the development of effective strategies to curb the proliferation of antibiotic resistance.
Collapse
Affiliation(s)
- Dengke Han
- Department of Laboratory Medicine, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China
| | - Suzhen Ma
- Department of Laboratory Medicine, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China
| | - Chenhong He
- Department of Emergency, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China
| | - Yuxing Yang
- Department of Laboratory Medicine, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China
| | - Peng Li
- Chinese PLA Center for Disease Control and Prevention, 20 DongDa Street, Fengtai District, Beijing, 100071, China
| | - Lanfen Lu
- Department of Laboratory Medicine, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China.
| |
Collapse
|
26
|
Naseef Pathoor N, Viswanathan A, Wadhwa G, Ganesh PS. Understanding the biofilm development of Acinetobacter baumannii and novel strategies to combat infection. APMIS 2024; 132:317-335. [PMID: 38444124 DOI: 10.1111/apm.13399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/22/2024] [Indexed: 03/07/2024]
Abstract
Acinetobacter baumannii (A. baumannii) is a Gram-negative, nonmotile, and aerobic bacillus emerged as a superbug, due to increasing the possibility of infection and accelerating rates of antimicrobial agents. It is recognized as a nosocomial pathogen due to its ability to form biofilms. These biofilms serve as a defensive barrier, increase antibiotic resistance, and make treatment more difficult. As a result, the current situation necessitates the rapid emergence of novel therapeutic approaches to ensure successful treatment outcomes. This review explores the intricate relationship between biofilm formation and antibiotic resistance in A. baumannii, emphasizing the role of key virulence factors and quorum sensing (QS) mechanisms that will lead to infections and facilitate insight into developing innovative method to control A. baumannii infections. Furthermore, the review article looks into promising approaches for preventing biofilm formation on medically important surfaces and potential therapeutic methods for eliminating preformed biofilms, which can address biofilm-associated A. baumannii infections. Modern advances in emerging therapeutic options such as antimicrobial peptide (AMPs), nanoparticles (NPs), bacteriophage therapy, photodynamic therapy (PDT), and other biofilm inhibitors can assist readers understand the current landscape and future prospects for effectively treating A. baumannii biofilm infections.
Collapse
Affiliation(s)
- Naji Naseef Pathoor
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, India
| | - Akshaya Viswanathan
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, India
| | - Gulshan Wadhwa
- Department of Biotechnology, Ministry of Science and Technology, New Delhi, India
| | - Pitchaipillai Sankar Ganesh
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, India
| |
Collapse
|
27
|
Macheras GA, Argyrou C, Tzefronis D, Milaras C, Tsivelekas K, Tsiamtsouris KG, Kateros K, Papadakis SA. Intraoperative calprotectin lateral flow immunoassay can assist decision-making between one- and two-stage revision total hip arthroplasty for patients with suspected periprosthetic joint infection. Bone Joint J 2024; 106-B:118-124. [PMID: 38688513 DOI: 10.1302/0301-620x.106b5.bjj-2023-0848.r1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Aims Accurate diagnosis of chronic periprosthetic joint infection (PJI) presents a significant challenge for hip surgeons. Preoperative diagnosis is not always easy to establish, making the intraoperative decision-making process crucial in deciding between one- and two-stage revision total hip arthroplasty (THA). Calprotectin is a promising point-of-care novel biomarker that has displayed high accuracy in detecting PJI. We aimed to evaluate the utility of intraoperative calprotectin lateral flow immunoassay (LFI) in THA patients with suspected chronic PJI. Methods The study included 48 THAs in 48 patients with a clinical suspicion of PJI, but who did not meet European Bone and Joint Infection Society (EBJIS) PJI criteria preoperatively, out of 105 patients undergoing revision THA at our institution for possible PJI between November 2020 and December 2022. Intraoperatively, synovial fluid calprotectin was measured with LFI. Cases with calprotectin levels ≥ 50 mg/l were considered infected and treated with two-stage revision THA; in negative cases, one-stage revision was performed. At least five tissue cultures were obtained; the implants removed were sent for sonication. Results Calprotectin was positive (≥ 50 mg/l) in 27 cases; out of these, 25 had positive tissue cultures and/or sonication. Calprotectin was negative in 21 cases. There was one false negative case, which had positive tissue cultures. Calprotectin showed an area under the curve of 0.917, sensitivity of 96.2%, specificity of 90.9%, positive predictive value of 92.6%, negative predictive value of 95.2%, positive likelihood ratio of 10.6, and negative likelihood ratio of 0.04. Overall, 45/48 patients were correctly diagnosed and treated by our algorithm, which included intraoperative calprotectin measurement. This yielded a 93.8% concordance with postoperatively assessed EBJIS criteria. Conclusion Calprotectin can be a valuable tool in facilitating the intraoperative decision-making process for cases in which chronic PJI is suspected and diagnosis cannot be established preoperatively.
Collapse
Affiliation(s)
- George A Macheras
- 7th Department of Orthopaedic Surgery, Henry Dunant Hospital Center, Athens, Greece
| | - Chrysoula Argyrou
- 4th Department of Orthopaedic Surgery and Traumatology, KAT Attica General Hospital, Athens, Greece
| | - Dimitrios Tzefronis
- 7th Department of Orthopaedic Surgery, Henry Dunant Hospital Center, Athens, Greece
| | - Christos Milaras
- 7th Department of Orthopaedic Surgery, Henry Dunant Hospital Center, Athens, Greece
| | - Konstantinos Tsivelekas
- 2nd Department of Orthopaedic Surgery and Traumatology, KAT Attica General Hospital, Athens, Greece
| | | | | | - Stamatios A Papadakis
- 2nd Department of Orthopaedic Surgery and Traumatology, KAT Attica General Hospital, Athens, Greece
| |
Collapse
|
28
|
Srivastava A, Verma N, Kumar V, Apoorva P, Agarwal V. Biofilm inhibition/eradication: exploring strategies and confronting challenges in combatting biofilm. Arch Microbiol 2024; 206:212. [PMID: 38616221 DOI: 10.1007/s00203-024-03938-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 04/16/2024]
Abstract
Biofilms are complex communities of microorganisms enclosed in a self-produced extracellular matrix, posing a significant threat to different sectors, including healthcare and industry. This review provides an overview of the challenges faced due to biofilm formation and different novel strategies that can combat biofilm formation. Bacteria inside the biofilm exhibit increased resistance against different antimicrobial agents, including conventional antibiotics, which can lead to severe problems in livestock and animals, including humans. In addition, biofilm formation also imposes heavy economic pressure on industries. Hence it becomes necessary to explore newer alternatives to eradicate biofilms effectively without applying selection pressure on the bacteria. Excessive usage of antibiotics may also lead to an increase in the number of resistant strains as bacteria employ an advanced antimicrobial resistance mechanism. This review provides insight into multifaceted technologies like quorum sensing inhibition, enzymes, antimicrobial peptides, bacteriophage, phytocompounds, and nanotechnology to neutralize biofilms without developing antimicrobial resistance (AMR). Furthermore, it will pave the way for developing newer therapeutic agents to deal with biofilms more efficiently.
Collapse
Affiliation(s)
- Anmol Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Nidhi Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Vivek Kumar
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Pragati Apoorva
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India
| | - Vishnu Agarwal
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, 211004, Uttar Pradesh, India.
| |
Collapse
|
29
|
Chen Q, Dong Z, Yao X, Sun H, Pan X, Liu J, Huang R. Bactericidal and biofilm eradication efficacy of a fluorinated benzimidazole derivative, TFBZ, against methicillin-resistant Staphylococcus aureus. Front Pharmacol 2024; 15:1342821. [PMID: 38659587 PMCID: PMC11039886 DOI: 10.3389/fphar.2024.1342821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major inducement of nosocomial infections and its biofilm formation render the high tolerance to conventional antibiotics, which highlights the requirement to develop new antimicrobial agents urgently. In this study, we identified a fluorinated benzimidazole derivative, TFBZ, with potent antibacterial efficacy toward planktonic MRSA (MIC = 4 μg/mL, MBC = 8 μg/mL) and its persistent biofilms (≥99%, MBEC = 8 μg/mL). TFBZ manifested significant irreversible time-dependent killing against MRSA as characterized by diminished cell viability, bacterial morphological change and protein leakage. Furthermore, the results from CBD devices, crystal violet assay in conjunction with live/dead staining and scanning electron microscopy confirmed that TFBZ was capable of eradicating preformed MRSA biofilms with high efficiency. Simultaneously, TFBZ reduced the bacterial invasiveness and exerted negligible hemolysis and cytotoxicity toward mammalian cells, which ensuring the robust therapeutic effect on mouse skin abscess model. The transcriptome profiling and quantitative RT-PCR revealed that a set of encoding genes associated with cell adhesion, biofilm formation, translation process, cell wall biosynthesis was consistently downregulated in MRSA biofilms upon exposure to TFBZ. In conclusion, TFBZ holds promise as a valuable candidate for therapeutic applications against MRSA chronic infections.
Collapse
Affiliation(s)
- Qian Chen
- The Modernization Engineering Technology Research Center of Ethnic Minority Medicine of Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhihui Dong
- The Modernization Engineering Technology Research Center of Ethnic Minority Medicine of Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Xuedi Yao
- The Modernization Engineering Technology Research Center of Ethnic Minority Medicine of Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Huan Sun
- The Modernization Engineering Technology Research Center of Ethnic Minority Medicine of Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Xin Pan
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Jikai Liu
- The Modernization Engineering Technology Research Center of Ethnic Minority Medicine of Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Rong Huang
- The Modernization Engineering Technology Research Center of Ethnic Minority Medicine of Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| |
Collapse
|
30
|
Iaconis A, De Plano LM, Caccamo A, Franco D, Conoci S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms 2024; 12:639. [PMID: 38674584 PMCID: PMC11052202 DOI: 10.3390/microorganisms12040639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Biofilm (BF) can give rise to systemic infections, prolonged hospitalization times, and, in the worst case, death. This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, the main phases of the life cycle of BF and maturation will be described to identify potential targets for anti-BF approaches. Then, an approach acting on bacterial adhesion, quorum sensing (QS), and the extracellular polymeric substance (EPS) matrix will be introduced and discussed. Finally, bacteriophage-mediated strategies will be presented as innovative approaches against BF inhibition/destruction.
Collapse
Affiliation(s)
- Antonella Iaconis
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
- URT Lab Sens Beyond Nano—CNR-DSFTM, Department of Physical Sciences and Technologies of Matter, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
31
|
Lories B, Belpaire TER, Smeets B, Steenackers HP. Competition quenching strategies reduce antibiotic tolerance in polymicrobial biofilms. NPJ Biofilms Microbiomes 2024; 10:23. [PMID: 38503782 PMCID: PMC10951329 DOI: 10.1038/s41522-024-00489-6] [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: 10/20/2022] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Bacteria typically live in dense communities where they are surrounded by other species and compete for a limited amount of resources. These competitive interactions can induce defensive responses that also protect against antimicrobials, potentially complicating the antimicrobial treatment of pathogens residing in polymicrobial consortia. Therefore, we evaluate the potential of alternative antivirulence strategies that quench this response to competition. We test three competition quenching approaches: (i) interference with the attack mechanism of surrounding competitors, (ii) inhibition of the stress response systems that detect competition, and (iii) reduction of the overall level of competition in the community by lowering the population density. We show that either strategy can prevent the induction of antimicrobial tolerance of Salmonella Typhimurium in response to competitors. Competition quenching strategies can thus reduce tolerance of pathogens residing in polymicrobial communities and could contribute to the improved eradication of these pathogens via traditional methods.
Collapse
Affiliation(s)
- Bram Lories
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Tom E R Belpaire
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
- Division of Mechatronics, Biostatistics, and Sensors (MeBioS), Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Bart Smeets
- Division of Mechatronics, Biostatistics, and Sensors (MeBioS), Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Hans P Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium.
| |
Collapse
|
32
|
Filipić B, Ušjak D, Rambaher MH, Oljacic S, Milenković MT. Evaluation of novel compounds as anti-bacterial or anti-virulence agents. Front Cell Infect Microbiol 2024; 14:1370062. [PMID: 38510964 PMCID: PMC10951914 DOI: 10.3389/fcimb.2024.1370062] [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: 01/13/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Antimicrobial resistance is a global threat, leading to an alarming increase in the prevalence of bacterial infections that can no longer be treated with available antibiotics. The World Health Organization estimates that by 2050 up to 10 million deaths per year could be associated with antimicrobial resistance, which would equal the annual number of cancer deaths worldwide. To overcome this emerging crisis, novel anti-bacterial compounds are urgently needed. There are two possible approaches in the fight against bacterial infections: a) targeting structures within bacterial cells, similar to existing antibiotics; and/or b) targeting virulence factors rather than bacterial growth. Here, for the first time, we provide a comprehensive overview of the key steps in the evaluation of potential new anti-bacterial and/or anti-virulence compounds. The methods described in this review include: a) in silico methods for the evaluation of novel compounds; b) anti-bacterial assays (MIC, MBC, Time-kill); b) anti-virulence assays (anti-biofilm, anti-quorum sensing, anti-adhesion); and c) evaluation of safety aspects (cytotoxicity assay and Ames test). Overall, we provide a detailed description of the methods that are an essential tool for chemists, computational chemists, microbiologists, and toxicologists in the evaluation of potential novel antimicrobial compounds. These methods are cost-effective and have high predictive value. They are widely used in preclinical studies to identify new molecular candidates, for further investigation in animal and human trials.
Collapse
Affiliation(s)
- Brankica Filipić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Dušan Ušjak
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Martina Hrast Rambaher
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Marina T. Milenković
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
33
|
Kan YC, Guo R, Xu Y, Han LY, Bu WH, Han LX, Chu JJ. Investigating the in vitro antibacterial efficacy of composite bone cement incorporating natural product-based monomers and gentamicin. J Orthop Surg Res 2024; 19:169. [PMID: 38448971 PMCID: PMC10918884 DOI: 10.1186/s13018-024-04646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
OBJECTIVE The objective of this study is to investigate the impact of four natural product extracts, namely, aloe-emodin, quercetin, curcumin, and tannic acid, on the in vitro bacteriostatic properties and biocompatibility of gentamicin-loaded bone cement and to establish an experimental groundwork supporting the clinical utility of antibiotic-loaded bone cements (ALBC). METHODS Based on the components, the bone cement samples were categorized as follows: the gentamicin combined with aloe-emodin group, the gentamicin combined with quercetin group, the gentamicin combined with curcumin group, the gentamicin combined with tannic acid group, the gentamicin group, the aloe-emodin group, the quercetin group, the curcumin group, and the tannic acid group. Using the disk diffusion test, we investigated the antibacterial properties of the bone cement material against Staphylococcus aureus (n = 4). We tested cell toxicity and proliferation using the cell counting kit-8 (CCK-8) and examined the biocompatibility of bone cement materials. RESULTS The combination of gentamicin with the four natural product extracts resulted in significantly larger diameters of inhibition zones compared to gentamicin alone, and the difference was statistically significant (P < 0.05). Except for the groups containing tannic acid, cells in all other groups showed good proliferation across varying time intervals without displaying significant cytotoxicity (P < 0.05). CONCLUSION In this study, aloe-emodin, quercetin, curcumin, and tannic acid were capable of enhancing the in vitro antibacterial performance of gentamicin-loaded bone cement against S. aureus. While the groups containing tannic acid displayed moderate cytotoxicity in in vitro cell culture, all other groups showed no discernible cytotoxic effects.
Collapse
Affiliation(s)
- Yu-Chen Kan
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital, Affiliated to Anhui Medical University, No. 246 of Heping Road, Yaohai District, Hefei, Anhui, 230011, China
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, Anhui, 230032, China
| | - Rui Guo
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital, Affiliated to Anhui Medical University, No. 246 of Heping Road, Yaohai District, Hefei, Anhui, 230011, China
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yang Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Lu-Yang Han
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital, Affiliated to Anhui Medical University, No. 246 of Heping Road, Yaohai District, Hefei, Anhui, 230011, China
| | - Wen-Han Bu
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital, Affiliated to Anhui Medical University, No. 246 of Heping Road, Yaohai District, Hefei, Anhui, 230011, China
| | - Long-Xu Han
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital, Affiliated to Anhui Medical University, No. 246 of Heping Road, Yaohai District, Hefei, Anhui, 230011, China
| | - Jian-Jun Chu
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital, Affiliated to Anhui Medical University, No. 246 of Heping Road, Yaohai District, Hefei, Anhui, 230011, China.
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
34
|
Yunus J, Wan Dagang WRZ, Jamaluddin H, Jemon K, Mohamad SE, Jonet MA. Bacterial biofilm growth and perturbation by serine protease from Bacillus sp. Arch Microbiol 2024; 206:138. [PMID: 38436775 DOI: 10.1007/s00203-024-03857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 03/05/2024]
Abstract
In nature, bacteria are ubiquitous and can be categorized as beneficial or harmless to humans, but most bacteria have one thing in common which is their ability to produce biofilm. Biofilm is encased within an extracellular polymeric substance (EPS) which provides resistance against antimicrobial agents. Protease enzymes have the potential to degrade or promote the growth of bacterial biofilms. In this study, the effects of a recombinant intracellular serine protease from Bacillus sp. (SPB) on biofilms from Staphylococcus aureus, Acinetobacter baumannii, and Pseudomonas aeruginosa were analyzed. SPB was purified using HisTrap HP column and concentrated using Amicon 30 ultra-centrifugal filter. SPB was added with varying enzyme activity and assay incubation period after biofilms were formed in 96-well plates. SPB was observed to have contrasting effects on different bacterial biofilms, where biofilm degradations were observed for both 7-day-old A. baumannii (37.26%) and S. aureus (71.51%) biofilms. Meanwhile, SPB promoted growth of P. aeruginosa biofilm up to 176.32%. Compatibility between protein components in S. aureus biofilm with SPB as well as a simpler membrane structure morphology led to higher biofilm degradation for S. aureus compared to A. baumannii. However, SPB promoted growth of P. aeruginosa biofilm due likely to its degrading protein factors that are responsible for biofilm detachment and dispersion, thus resulting in more multi-layered biofilm formation. Commercial protease Savinase which was used as a comparison showed degradation for all three bacterial biofilms. The results obtained are unique and will expand our understanding on the effects that bacterial proteases have toward biofilms.
Collapse
Affiliation(s)
- Julia Yunus
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310, Skudai, Johor, Malaysia
| | - Wan Rosmiza Zana Wan Dagang
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310, Skudai, Johor, Malaysia.
| | - Haryati Jamaluddin
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310, Skudai, Johor, Malaysia.
| | - Khairunadwa Jemon
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310, Skudai, Johor, Malaysia
| | - Shaza Eva Mohamad
- Department of Environmental Engineering and Green Technology (EGT), Malaysia Japan International Institute of Technology (MJIIT), Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia
| | - Mohd Anuar Jonet
- Structural And Applied Genomics Centre, Malaysia Genome and Vaccine Institute (MGVI), National Institute of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
35
|
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
|
36
|
Surekha S, Lamiyan AK, Gupta V. Antibiotic Resistant Biofilms and the Quest for Novel Therapeutic Strategies. Indian J Microbiol 2024; 64:20-35. [PMID: 38468748 PMCID: PMC10924852 DOI: 10.1007/s12088-023-01138-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/03/2023] [Indexed: 03/13/2024] Open
Abstract
Antimicrobial resistance (AMR) is one of the major leading causes of death around the globe. Present treatment pipelines are insufficient to overcome the critical situation. Prominent biofilm forming human pathogens which can thrive in infection sites using adaptive features results in biofilm persistence. Considering the present scenario, prudential investigations into the mechanisms of resistance target them to improve antibiotic efficacy is required. Regarding this, developing newer and effective treatment options using edge cutting technologies in medical research is the need of time. The reasons underlying the adaptive features in biofilm persistence have been centred on different metabolic and physiological aspects. The high tolerance levels against antibiotics direct researchers to search for novel bioactive molecules that can help combat the problem. In view of this, the present review outlines the focuses on an opportunity of different strategies which are in testing pipeline can thus be developed into products ready to use.
Collapse
Affiliation(s)
- Saumya Surekha
- Department of Biochemistry, Panjab University, Chandigarh, India
| | | | - Varsha Gupta
- GMCH: Government Medical College and Hospital, Chandigarh, India
| |
Collapse
|
37
|
Confessor MVA, Agreles MAA, Campos LADA, Silva Neto AF, Borges JC, Martins RM, Scavuzzi AML, Lopes ACS, Kretzschmar EADM, Cavalcanti IMF. Olive oil nanoemulsion containing curcumin: antimicrobial agent against multidrug-resistant bacteria. Appl Microbiol Biotechnol 2024; 108:241. [PMID: 38413482 PMCID: PMC10899360 DOI: 10.1007/s00253-024-13057-x] [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: 07/22/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
The present work aimed to develop, characterize, and evaluate the antibacterial and antibiofilm activity of two nanoemulsions (NEs) containing 500 µg/mL of curcumin from Curcuma longa (CUR). These NEs, produced with heating, contain olive oil (5%) and the surfactants tween 80 (5%) and span 80 (2.5%), water q.s. 100 mL, and were stable for 120 days. NE-2-CUR presented Ø of 165.40 ± 2.56 nm, PDI of 0.254, ζ of - 33.20 ± 1.35 mV, pH of 6.49, and Entrapment Drug Efficiency (EE) of 99%. The NE-4-CUR showed a Ø of 105.70 ± 4.13 nm, PDI of 0.459, ζ of - 32.10 ± 1.45 mV, pH of 6.40 and EE of 99.29%. Structural characterization was performed using DRX and FTIR, thermal characterization using DSC and TG, and morphological characterization using SEM, suggesting that there is no significant change in the CUR present in the NEs and that they remain stable. The MIC was performed by the broth microdilution method for nine gram-positive and gram-negative bacteria, as well as Klebsiella pneumoniae clinical isolates resistant to antibiotics and biofilm and efflux pump producers. The NEs mostly showed a bacteriostatic profile. The MIC varied between 125 and 250 µg/mL. The most sensitive bacteria were Staphylococcus aureus and Enterococcus faecalis, for which NE-2-CUR showed a MIC of 125 µg/mL. The NEs and ceftazidime (CAZ) interaction was also evaluated against the K. pneumoniae resistant clinical isolates using the Checkerboard method. NE-2-CUR and NE-4-CUR showed a synergistic or additive profile; there was a reduction in CAZ MICs between 256 times (K26-A2) and 2 times (K29-A2). Furthermore, the NEs inhibited these isolates biofilms formation. The NEs showed a MBIC ranging from 15.625 to 250 µg/mL. Thus, the NEs showed physicochemical characteristics suitable for future clinical trials, enhancing the CAZ antibacterial and antibiofilm activity, thus becoming a promising strategy for the treatment of bacterial infections caused by multidrug-resistant K. pneumoniae. KEY POINTS: • The NEs showed physicochemical characteristics suitable for future clinical trials. • The NEs showed a synergistic/additive profile, when associated with ceftazidime. • The NEs inhibited biofilm formation of clinical isolates.
Collapse
Affiliation(s)
- Maine Virgínia Alves Confessor
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Prof. Moraes Rego Avenue, 1235, Cidade Universitária, CEP, Recife, Pernambuco, 50670-901, Brazil.
- University Center UNIFACISA, Manoel Cardoso Palhano, 124-152, Itararé, CEP, Campina Grande, Paraiba, 58408-326, Brazil.
| | - Maria Anndressa Alves Agreles
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Prof. Moraes Rego Avenue, 1235, Cidade Universitária, CEP, Recife, Pernambuco, 50670-901, Brazil
| | - Luís André de Almeida Campos
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Prof. Moraes Rego Avenue, 1235, Cidade Universitária, CEP, Recife, Pernambuco, 50670-901, Brazil
| | - Azael Francisco Silva Neto
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Prof. Moraes Rego Avenue, 1235, Cidade Universitária, CEP, Recife, Pernambuco, 50670-901, Brazil
| | - Joyce Cordeiro Borges
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Prof. Moraes Rego Avenue, 1235, Cidade Universitária, CEP, Recife, Pernambuco, 50670-901, Brazil
| | - Rodrigo Molina Martins
- University Center UNIFACISA, Manoel Cardoso Palhano, 124-152, Itararé, CEP, Campina Grande, Paraiba, 58408-326, Brazil
| | | | - Ana Catarina Souza Lopes
- Department of Tropical Medicine, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | | | - Isabella Macário Ferro Cavalcanti
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Prof. Moraes Rego Avenue, 1235, Cidade Universitária, CEP, Recife, Pernambuco, 50670-901, Brazil
- Laboratory of Microbiology and Immunology, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão, Pernambuco, Brazil
| |
Collapse
|
38
|
Bilgin M, Dosler S, Otuk G. Antibiotic adjuvant activities of quorum sensing signal molecules DSF and BDSF against mature biofilms of Staphylococci. J Chemother 2024; 36:11-23. [PMID: 37873740 DOI: 10.1080/1120009x.2023.2270743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
Among promising antibiofilm compounds, quorum-sensing (QS) molecules that regulate biological processes such as biofilm formation and intra- or interspecies communication appear to be good candidates. The invitro antibiotic-adjuvant effects of QS molecules diffusible signal factor (DSF) and B. cenocepacia producing-DSF (BDSF) were investigated against mature Staphylococcal biofilms. Broth microdilution methods were used for the determinations of MIC, MBC, MBIC, and MBEC, and bactericidal activities were determined by TKC method. The lowest MICs were obtained with ciprofloxacin and gentamicin, and MBECs with ciprofloxacin. DSF and BDSF at 0.5 µM decreased the MICs as 2-8, and 2-32 fold, respectively. In TKC studies, -cidal activities were achieved by BDSF + gentamycin, or ciprofloxacin, and DSF + daptomycin, vancomycin, meropenem or gentamycin combinations. Synergistic effects were generally obtained with BDSF + gentamicin combinations, followed by DSF + daptomycin against most S. aureus; while BDSF + gentamicin or ciprofloxacin, and DSF + vancomycin or meropenem were synergist against some S. epidermidis biofilms. Also, the antagonist effects were observed with BDSF + meropenem or ciprofloxacin against each MSSE and MSSA. It is estimated that these QS molecules, although it was strain dependent, generally enhanced the antibiotic activity, and would be a new and effective treatment strategy for biofilm control, either alone or as an antibiotic adjuvant.
Collapse
Affiliation(s)
- Merve Bilgin
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkiye
- Department of Pharmaceutical Microbiology, Istanbul Health & Technology University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Sibel Dosler
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Gulten Otuk
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| |
Collapse
|
39
|
Dzięgielewska M, Bartoszewicz M, Książczyk M, Dudek B, Brożyna M, Szymczyk-Ziółkowska P, Gruber P, Pawlak J, Kozłowska W, Zielińska S, Fischer J, Woytoń A, Junka A. Abietic Acid as a Novel Agent against Ocular Biofilms: An In Vitro and Preliminary In Vivo Investigation. Int J Mol Sci 2024; 25:1528. [PMID: 38338807 PMCID: PMC10855443 DOI: 10.3390/ijms25031528] [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: 12/08/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Biofilm-related ocular infections can lead to vision loss and are difficult to treat with antibiotics due to challenges with application and increasing microbial resistance. In turn, the design and testing of new synthetic drugs is a time- and cost-consuming process. Therefore, in this work, for the first time, we assessed the in vitro efficacy of the plant-based abietic acid molecule, both alone and when introduced to a polymeric cellulose carrier, against biofilms formed by Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans in standard laboratory settings as well as in a self-designed setting using the topologically challenging surface of the artificial eye. These analyses were performed using the standard microdilution method, the biofilm-oriented antiseptic test (BOAT), a modified disk-diffusion method, and eyeball models. Additionally, we assessed the cytotoxicity of abietic acid against eukaryotic cell lines and its anti-staphylococcal efficacy in an in vivo model using Galleria mellonella larvae. We found that abietic acid was more effective against Staphylococcus than Pseudomonas (from two to four times, depending on the test applied) and that it was generally more effective against the tested bacteria (up to four times) than against the fungus C. albicans at concentrations non-cytotoxic to the eukaryotic cell lines and to G. mellonella (256 and 512 µg/mL, respectively). In the in vivo infection model, abietic acid effectively prevented the spread of staphylococcus throughout the larvae organisms, decreasing their lethality by up to 50%. These initial results obtained indicate promising features of abietic acid, which may potentially be applied to treat ocular infections caused by pathogenic biofilms, with higher efficiency manifested against bacterial than fungal biofilms.
Collapse
Affiliation(s)
| | - Marzenna Bartoszewicz
- Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, 50-367 Wroclaw, Poland;
| | - Marta Książczyk
- Department of Microbiology, Institute of Genetics and Microbiology, University of Wrocław, 51-148 Wroclaw, Poland;
| | - Bartłomiej Dudek
- Platform for Unique Model Application, Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, 50-367 Wroclaw, Poland; (M.B.); (A.W.)
| | - Malwina Brożyna
- Platform for Unique Model Application, Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, 50-367 Wroclaw, Poland; (M.B.); (A.W.)
| | - Patrycja Szymczyk-Ziółkowska
- Center for Advanced Manufacturing Technologies (CAMT/FPC), Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Łukasiewicza 5, 50-371 Wroclaw, Poland; (P.S.-Z.); (P.G.)
| | - Piotr Gruber
- Center for Advanced Manufacturing Technologies (CAMT/FPC), Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Łukasiewicza 5, 50-371 Wroclaw, Poland; (P.S.-Z.); (P.G.)
| | - Jacek Pawlak
- Medical Department, Lazarski University, 02-662 Warsaw, Poland;
| | - Weronika Kozłowska
- Department of Pharmaceutical Biology and Biotechnology, Division of Pharmaceutical Biotechnology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (W.K.); (S.Z.)
| | - Sylwia Zielińska
- Department of Pharmaceutical Biology and Biotechnology, Division of Pharmaceutical Biotechnology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (W.K.); (S.Z.)
| | - Jędrzej Fischer
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Aleksandra Woytoń
- Platform for Unique Model Application, Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, 50-367 Wroclaw, Poland; (M.B.); (A.W.)
| | - Adam Junka
- Platform for Unique Model Application, Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw, 50-367 Wroclaw, Poland; (M.B.); (A.W.)
| |
Collapse
|
40
|
Piątczak E, Kolniak-Ostek J, Gonciarz W, Lisiecki P, Kalinowska-Lis U, Szemraj M, Chmiela M, Zielińska S. The Effect of Salvia tomentosa Miller Extracts, Rich in Rosmarinic, Salvianolic and Lithospermic Acids, on Bacteria Causing Opportunistic Infections. Molecules 2024; 29:590. [PMID: 38338335 PMCID: PMC10856039 DOI: 10.3390/molecules29030590] [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: 12/30/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Methanolic-aqueous extracts of Salvia tomentosa Miller roots, aerial parts, and inflorescences were examined for their content of polyphenolic derivatives and the antimicrobial and cytotoxic effect. In the polyphenolic-rich profile, rosmarinic, salvianolic, and lithospermic acids along with various derivatives were predominant. A total of twenty phenolic compounds were identified using the UPLC/DAD/qTOF-MS technique. These were caffeic acid, rosmarinic acid derivatives, lithospermic acid derivatives, salvianolic acids B, F, and K derivatives, as well as sagerinic acid, although rosmarinic acid (426-525 mg/100 g of dry weight-D.W.) and salvianolic acid B (83-346.5 mg/100 g D.W.) were significantly predominant in the metabolic profile. Strong antibacterial activity of S. tomentosa extracts was observed against Staphylococcus epidermidis (MIC/MBC = 0.625 mg/mL) and Bacillus cereus (MIC = 0.312-1.25 mg/mL). The extracts showed low cytotoxicity towards the reference murine fibroblasts L929 and strong cytotoxicity to human AGS gastric adenocarcinoma epithelial cells in the MTT reduction assay. The observed cytotoxic effect in cancer cells was strongest for the roots of 2-year-old plant extracts.
Collapse
Affiliation(s)
- Ewelina Piątczak
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Joanna Kolniak-Ostek
- Department of Fruit, Vegetable and Plant Nutraceutical Technology, Wrocław University of Environmental and Life Sciences, Chełmońskiego 37, 51-630 Wroclaw, Poland;
| | - Weronika Gonciarz
- Department of Immunology and Infectious Biology, Faculty of Biology and Environment Protections, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (W.G.); (M.C.)
| | - Paweł Lisiecki
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland; (P.L.); (M.S.)
| | - Urszula Kalinowska-Lis
- Department of Cosmetic Raw Materials Chemistry, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Magdalena Szemraj
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland; (P.L.); (M.S.)
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Faculty of Biology and Environment Protections, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (W.G.); (M.C.)
| | - Sylwia Zielińska
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| |
Collapse
|
41
|
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
|
42
|
Chung FY, Lin YZ, Huang CR, Huang KW, Chen YF. Crosslinking kiwifruit-derived DNA with natural aromatic aldehydes generates membranolytic antibacterial nanogels. Int J Biol Macromol 2024; 255:127947. [PMID: 37951422 DOI: 10.1016/j.ijbiomac.2023.127947] [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: 06/16/2023] [Revised: 10/14/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023]
Abstract
Improper use of antibiotics has led to the global rise of drug-resistant biofilm bacteria. Thus, researchers have been increasingly interested in green materials that are highly biocompatible and have low toxicity. Here, nanogels (NGs) with imine bonds were synthesized by crosslinking kiwifruit-derived DNA's primary amine and aromatic aldehydes (cuminaldehyde, p-anisaldehyde, or vanillin) under water-in-hexane emulsion processes. Transmission electron microscope showed that the NGs had spherical geometry with an average particle size ranging from 40 to 140 nm and that the zeta potential indicated a negative charge. Additionally, the DNA-aromatic aldehyde NGs showed low cytotoxicity toward normal cell organoids and human RBCs in cell viability tests. These NGs were also tested against four pathogenic bacteria for various assays. DNA-vanillin (DNA-VA) NGs exhibited significant antibacterial effects against bacteria with very low inhibitory concentrations as seen in a minimum inhibitory concentration assay. Scanning electron microscope observation revealed that the bacteria were deformed, and immunoblotting detected intracellular groEL protein expression. In agreement with these results, DNA-aromatic aldehyde NGs successfully protected C. elegans from P. aeruginosa-induced lethality. These DNA NGs provided a multivalent 3D space for antibacterial aromatic aldehydes to tether, enhancing their interaction with the bacterial wall. These results offer a new direction for the development of novel antibiotics in the future.
Collapse
Affiliation(s)
- Fang-Yu Chung
- Master Program in Biomedicine, National Taitung University, No. 684, Section 1, Zhonghua Rd., Taitung 95092, Taiwan
| | - Yi-Zhen Lin
- Master Program in Biomedicine, National Taitung University, No. 684, Section 1, Zhonghua Rd., Taitung 95092, Taiwan
| | - Cheng-Rung Huang
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, No. 1, University Rd., East Dist., Tainan 70101, Taiwan
| | - Kuan-Wen Huang
- Master Program in Biomedicine, National Taitung University, No. 684, Section 1, Zhonghua Rd., Taitung 95092, Taiwan
| | - Yu-Fon Chen
- Master Program in Biomedicine, National Taitung University, No. 684, Section 1, Zhonghua Rd., Taitung 95092, Taiwan.
| |
Collapse
|
43
|
Blanco-Cabra N, Alcàcer-Almansa J, Admella J, Arévalo-Jaimes BV, Torrents E. Nanomedicine against biofilm infections: A roadmap of challenges and limitations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1944. [PMID: 38403876 DOI: 10.1002/wnan.1944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/28/2023] [Accepted: 01/27/2024] [Indexed: 02/27/2024]
Abstract
Microbial biofilms are complex three-dimensional structures where sessile microbes are embedded in a polymeric extracellular matrix. Their resistance toward the host immune system as well as to a diverse range of antimicrobial treatments poses a serious health and development threat, being in the top 10 global public health threats declared by the World Health Organization. In an effort to combat biofilm-related microbial infections, several strategies have been developed to independently eliminate biofilms or to complement conventional antibiotic therapies. However, their limitations leave room for other treatment alternatives, where the application of nanotechnology to biofilm eradication has gained significant relevance in recent years. Their small size, penetration efficiency, and the design flexibility that they present makes them a promising alternative for biofilm infection treatment, although they also present set-backs. This review aims to describe the main possibilities and limitations of nanomedicine against biofilms, while covering the main aspects of biofilm formation and study, and the current therapies for biofilm treatment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Núria Blanco-Cabra
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Júlia Alcàcer-Almansa
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joana Admella
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Betsy Verónica Arévalo-Jaimes
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
44
|
Kumar V, Yasmeen N, Pandey A, Ahmad Chaudhary A, Alawam AS, Ahmad Rudayni H, Islam A, Lakhawat SS, Sharma PK, Shahid M. Antibiotic adjuvants: synergistic tool to combat multi-drug resistant pathogens. Front Cell Infect Microbiol 2023; 13:1293633. [PMID: 38179424 PMCID: PMC10765517 DOI: 10.3389/fcimb.2023.1293633] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024] Open
Abstract
The rise of multi-drug resistant (MDR) pathogens poses a significant challenge to the field of infectious disease treatment. To overcome this problem, novel strategies are being explored to enhance the effectiveness of antibiotics. Antibiotic adjuvants have emerged as a promising approach to combat MDR pathogens by acting synergistically with antibiotics. This review focuses on the role of antibiotic adjuvants as a synergistic tool in the fight against MDR pathogens. Adjuvants refer to compounds or agents that enhance the activity of antibiotics, either by potentiating their effects or by targeting the mechanisms of antibiotic resistance. The utilization of antibiotic adjuvants offers several advantages. Firstly, they can restore the effectiveness of existing antibiotics against resistant strains. Adjuvants can inhibit the mechanisms that confer resistance, making the pathogens susceptible to the action of antibiotics. Secondly, adjuvants can enhance the activity of antibiotics by improving their penetration into bacterial cells, increasing their stability, or inhibiting efflux pumps that expel antibiotics from bacterial cells. Various types of antibiotic adjuvants have been investigated, including efflux pump inhibitors, resistance-modifying agents, and compounds that disrupt bacterial biofilms. These adjuvants can act synergistically with antibiotics, resulting in increased antibacterial activity and overcoming resistance mechanisms. In conclusion, antibiotic adjuvants have the potential to revolutionize the treatment of MDR pathogens. By enhancing the efficacy of antibiotics, adjuvants offer a promising strategy to combat the growing threat of antibiotic resistance. Further research and development in this field are crucial to harness the full potential of antibiotic adjuvants and bring them closer to clinical application.
Collapse
Affiliation(s)
- Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
- Amity Institute of Pharmacy, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Nusrath Yasmeen
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Aishwarya Pandey
- INRS, Eau Terre Environnement Research Centre, Québec, QC, Canada
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Abdullah S. Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Hassan Ahmad Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Sudarshan S. Lakhawat
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Pushpender K. Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
45
|
Kornicka A, Balewski Ł, Lahutta M, Kokoszka J. Umbelliferone and Its Synthetic Derivatives as Suitable Molecules for the Development of Agents with Biological Activities: A Review of Their Pharmacological and Therapeutic Potential. Pharmaceuticals (Basel) 2023; 16:1732. [PMID: 38139858 PMCID: PMC10747342 DOI: 10.3390/ph16121732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Umbelliferone (UMB), known as 7-hydroxycoumarin, hydrangine, or skimmetine, is a naturally occurring coumarin in the plant kingdom, mainly from the Umbelliferae family that possesses a wide variety of pharmacological properties. In addition, the use of nanoparticles containing umbelliferone may improve anti-inflammatory or anticancer therapy. Also, its derivatives are endowed with great potential for therapeutic applications due to their broad spectrum of biological activities such as anti-inflammatory, antioxidant, neuroprotective, antipsychotic, antiepileptic, antidiabetic, antimicrobial, antiviral, and antiproliferative effects. Moreover, 7-hydroxycoumarin ligands have been implemented to develop 7-hydroxycoumarin-based metal complexes with improved pharmacological activity. Besides therapeutic applications, umbelliferone analogues have been designed as fluorescent probes for the detection of biologically important species, such as enzymes, lysosomes, and endosomes, or for monitoring cell processes and protein functions as well various diseases caused by an excess of hydrogen peroxide. Furthermore, 7-hydroxy-based chemosensors may serve as a highly selective tool for Al3+ and Hg2+ detection in biological systems. This review is devoted to a summary of the research on umbelliferone and its synthetic derivatives in terms of biological and pharmaceutical properties, especially those reported in the literature during the period of 2017-2023. Future potential applications of umbelliferone and its synthetic derivatives are presented.
Collapse
Affiliation(s)
- Anita Kornicka
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland; (Ł.B.); (M.L.); (J.K.)
| | | | | | | |
Collapse
|
46
|
Brindhadevi K, Hoang Le Q, Salmen SH, Karuppusamy I, Pugazhendhi A. In vitro biofilm inhibition efficacy of Aerva lanata flower extract against Gram negative and Gram-positive biofilm forming bacteria and toxicity analysis using Artemia salina. ENVIRONMENTAL RESEARCH 2023; 238:117118. [PMID: 37704075 DOI: 10.1016/j.envres.2023.117118] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/24/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
A biofilm consists of Gram positive and Gram-negative bacteria enclosed in a matrix. Industrial biofouling is caused by biofilms, which can exhibit antimicrobial resistance during infections. Many biofilm studies find that nearly all biofilm communities consist of Gram positive and Gram-negative bacteria. It is therefore necessary to better understand the conserved themes in biofilm formation to develop therapeutics based on biofilm formation. Plant extracts can effectively combat pathogenic bacterial biofilms. This study evaluated the antibacterial and antibiofilm activity of Aerva lanata flower extract against Staphylococcus aureus and Pseudomonas aeruginosa. Methanol extract of dried A. lanata flower was tested against S. aureus and P. aeruginosa to determine the antibacterial activity (10, 25, 50, 75, 100 μg/mL) resulted in a maximum of 0.5-1 log reduction and 2 log reduction in comparison to the control or untreated bacterial cells respectively. A. lanata showed maximum biofilm inhibition up to 1.5-fold and 1-fold against P. aeruginosa and S. aureus. Light microscopic analysis of biofilm treated with A. lanata extract showed efficient distortion of the biofilm matrix. Further, the in vivo analysis of A. lanata in the Artemia salina brine shrimp model showed >50% survival and thus proving the efficacy of A. lanata extract in rescuing the brine shrimps against P. aeruginosa and S. aureus infection.
Collapse
Affiliation(s)
- Kathirvel Brindhadevi
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Viet Nam; Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam.
| | - Quynh Hoang Le
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Viet Nam; Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam
| | - Saleh H Salmen
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| | - Indira Karuppusamy
- Research Center for Strategic Materials, Corrosion Resistant Steel Group, National Institute for Materials Science (NIMS), Tsukuba, Japan
| | - Arivalagan Pugazhendhi
- School of Engineering, Lebanese American University, Byblos, Lebanon; Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
| |
Collapse
|
47
|
Ghanipour F, Nazari R, Aghaei SS, Jafari P. Effect of lipopeptide extracted from Bacillus licheniformis on the expression of bap and luxI genes in multi-drug-resistant Acinetobacter baumannii and Pseudomonas aeruginosa. Amino Acids 2023; 55:1891-1907. [PMID: 37907777 DOI: 10.1007/s00726-023-03346-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
Recently, opportunistic pathogens like Acinetobacter baumannii and Pseudomonas aeruginosa have caused concern due to their ability to cause antibiotic resistance in weakened immune systems. As a result, researchers are always seeking efficient antimicrobial agents to tackle this issue. The hypothesis of the recent study was that probiotic products derived from bacteria would be effective in reducing drug resistance in other bacteria. This research aimed to investigate the antimicrobial properties of probiotic products from various bacterial strains, including Lactobacillus rhamnosus, Pediococcus acidilactisi, Bacillus coagulans, Bacillus subtilis, and Bacillus licheniformis. These were tested against multi-drug-resistant (MDR) standard strains A. baumannii and P. aeruginosa. B. licheniformis was found to be the most effective probiotic strain, possessing the LanA and LanM lantibiotic genes. The lipopeptide nature of the probiotic product was confirmed through high-performance liquid chromatography (HPLC) and Fourier-transform infrared spectroscopy (FTIR) techniques. The anti-biofilm and antimicrobial properties of this probiotic were measured using an SEM electron microscope and minimum inhibitory concentration (MIC) test. Real-time PCR (qPCR) was used to compare the expression of bap and luxI genes, which are considered virulence factors of drug-resistant bacteria, before and after treatment with antimicrobial agents. The MIC results showed that the probiotic product prevented the growth of bacteria at lower concentrations compared to antibiotics. In addition, the ΔΔCqs indicated that gene expression was significantly down-regulated following treatment with the obtained probiotic product. It was found that B. licheniformis probiotic products could reduce drug resistance in other bacteria, making it a potential solution to antibiotic resistance.
Collapse
Affiliation(s)
- Farangis Ghanipour
- Department of Microbiology, Faculty of Basic Sciences, Qom Branch, Islamic Azad University, 15 Khordad Boulevard, Qom, Iran
| | - Razieh Nazari
- Department of Microbiology, Faculty of Basic Sciences, Qom Branch, Islamic Azad University, 15 Khordad Boulevard, Qom, Iran.
| | - Seyed Soheil Aghaei
- Department of Microbiology, Faculty of Basic Sciences, Qom Branch, Islamic Azad University, 15 Khordad Boulevard, Qom, Iran
| | - Parvaneh Jafari
- Department of Microbiology, Faculty of Basic Sciences, Arak Branch, Islamic Azad University, Arak, 3749113191, Iran
| |
Collapse
|
48
|
Kumar A, Kumar RR, Chaturvedi V, Kayastha AM. α-Amylase purified and characterized from fenugreek (Trigonella foenum-graecum) showed substantial anti-biofilm activity against Staphylococcus aureus MTCC740. Int J Biol Macromol 2023; 252:126442. [PMID: 37611683 DOI: 10.1016/j.ijbiomac.2023.126442] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Starch hydrolyzing α-amylase from germinated fenugreek (Trigonella foenum-graecum) has been purified 104-fold to apparent electrophoretic homogeneity with a final specific activity of 297.5 units/mg. SDS-PAGE of the final preparation revealed a single protein band of 47.5 kDa, supported by LC/MS analysis and size-exclusion chromatography on the Superdex 200 (ÄKTA-FPLC). α-Amylase exhibited maximum activity at pH 5.5. An activation energy (Ea) of 9.12 kcal/mol was found to exist in the temperature range of 20 to 90 °C. When substrate concentrations were evaluated between 0.5 and 10 mg/mL, the Km and Vmax values for starch were observed to be 1.12 mg/mL and 384.14 μmol/min/mg, respectively. The major substrate starch exhibited high specificity for fenugreek α-amylase. In the presence of EDTA (5 mM), the activity was lost, however, it could be largely reversed with the addition of calcium. Furthermore, an effort was made to assess the ability of fenugreek seed-derived partially purified (DEAE-cellulose enzyme) and purified α-amylase to disperse inside 48 h-old biofilms of Staphylococcus aureus MTCC740. The outcomes clearly demonstrated that the purified and partially purified α-amylase both exhibited strong biofilm dispersion activity.
Collapse
Affiliation(s)
- Avinash Kumar
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Ravi Ranjan Kumar
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Venkatesh Chaturvedi
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Arvind M Kayastha
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
49
|
Kuppusamy KM, Selvaraj S, Singaravelu P, John CM, Racheal K, Varghese K, Kaliyamoorthy D, Perumal E, Gunasekaran K. Anti-microbial and anti-cancer efficacy of acetone extract of Rosa chinensis against resistant strain and lung cancer cell line. BMC Complement Med Ther 2023; 23:406. [PMID: 37950173 PMCID: PMC10636979 DOI: 10.1186/s12906-023-04222-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Screening of herbal plants for various therapeutic properties is the hour as it shows promising activity. Scientific evidence of the pharmacological activity of the plant strengthens the traditional application of plants. METHODS Rose flowers (Rosa chinensis) were procured and grounded into a coarse powder. The DNA was isolated from rose flower and molecular identification was performed by rbcL-BF and rbcL-724R primers. Antibacterial activity was evaluated by using disc and agar diffusion methods and the anti-cancer effect of the rose flower extract (RE) was examined using MTT assay in lung cancer cell line. The mechanism of cell death induced by RE was qualitatively measured using Acridine orange/Ethidium bromide staining and Hoechst staining. GC-MS analysis was performed using GC-MS-5975C. RESULT The RE showed potent antimicrobial activity against various ATCC cultures. The rose extract strongly inhibits the growth of ESBL resistant organism along with inhibition of biofilm formation in the ESBL resistant organism. The extract caused apoptotic and necrotic cell death in lung cancer cells. GC-MS analysis demonstrated the presence of several biologically active compounds such as Clindamycin, Phytol, Octanoic acid, and Stigmasterol which might be the reason for the therapeutic properties of the plant. CONCLUSION This study shows the antimicrobial and biofilm inhibition activity against the clinical isolates of Klebsiella pneumonia. The study shows the cytotoxic and apoptotic activity in A549 cancer cell line. Thus, the plant may act as a potent antimicrobial drug against resistant strains.
Collapse
Affiliation(s)
- Kalaivani Madhavaram Kuppusamy
- Research Centre for Cellular Genomics and Cancer Research, Sree Balaji Medical College and Hospital, Chennai, 600044, India
| | - Sivakumar Selvaraj
- Molecular Biology section, Consultant Molecular Biologist, Medall Healthcare Private Limited, Chennai, India
| | - Pujithaa Singaravelu
- Research Centre for Cellular Genomics and Cancer Research, Sree Balaji Medical College and Hospital, Chennai, 600044, India
| | - Cordelia Mano John
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Kalaiselvan Racheal
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Keziaann Varghese
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Dinesh Kaliyamoorthy
- Department of Microbiology, Sree Balaji Medical College and Hospital, Chennai, 600044, India
| | - Elumalai Perumal
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Krishnamoorthy Gunasekaran
- Department of Medical Biochemistry, College of Health Sciences, Dambi Dollo University, KelamWelega Zone, Dembidolo, P.O. Box: 360, Oromia Region, Ethiopia.
| |
Collapse
|
50
|
Shaygankho N, Jahangiri A, Rasooli I. Passive immunization with anti-FimA egg yolk antibodies (IgYs) mitigate Acinetobacter baumannii pneumonia in mice. Biomed Pharmacother 2023; 167:115583. [PMID: 37774673 DOI: 10.1016/j.biopha.2023.115583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
Acinetobacter baumannii is a formidable pathogen, characterized by high mortality rates and pan-drug-resistant strains. Current commercial antibiotics lack efficacy against drug-resistant variants, necessitating the search for alternative treatments. This study investigates the potential of egg yolk immunoglobulin (IgY) as a cost-effective biomolecule for passive protection against A. baumannii pneumonia. FimA (ABAYE2132), a key virulence factor involved in biofilm development and lung cell adherence, emerges as a promising antigen for triggering protective IgY production. Recombinant FimA was expressed, purified, and used for intramuscular immunization of laying White Leghorn hens. IgY antibodies were subsequently extracted from egg yolks, with their reactivity assessed through indirect ELISA. Neutropenic mice received intranasal administration of IgYs one hour prior to the challenge with a clinical A. baumannii isolate (10 ×LD50). The specific anti-FimA IgYs detected recombinant FimA and provided 100% protection against bacterial infection, while non-specific IgYs prolonged survival for up to 72 h. In contrast, control mice succumbed to infection within 24 h. Analysis of bacterial loads in lungs and spleens after 16 h reveals the following order: control > non-specific IgY > anti-FimA IgY. These findings highlight FimA as a suitable antigen for the development of protective IgYs against A. baumannii.
Collapse
Affiliation(s)
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems biology and poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran; Molecular Microbiology Research Center and Department of Biology, Shahed University, Tehran, Iran.
| |
Collapse
|