1
|
Asmamaw D, Mwangi J, Michira BB, Khalid M, Prateeksha, Yang M, Lu Q, Yi W, Thuku RC, Duan Z, Lai R. Broad-Spectrum Antimicrobial Efficacy of Cyclic Antimicrobial Peptide Against Multidrug-Resistant Staphylococcus aureus Infections. ACS Med Chem Lett 2025; 16:1114-1123. [PMID: 40529091 PMCID: PMC12169473 DOI: 10.1021/acsmedchemlett.5c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 06/20/2025] Open
Abstract
The increasing emergence and spread of multidrug-resistant (MDR) bacteria have intensified the search for novel antimicrobial peptides (AMPs). Here, we developed SAP 2.8 a synthetic amphipathic helical peptide, with the sequence "RCWKRWWRWWKRCWR", that demonstrates potent antibacterial activity, antibiofilm properties, and a well-characterized mechanism of action. SAP 2.8 peptide demonstrated a remarkable antibacterial effect against MDR Staphylococcus aureus (S. aureus) and Methicillin-resistant Staphylococcus aureus (MRSA) clinical isolates, with minimum inhibitory concentrations (MICs) ranging from 1.25 to 2.5 μg/mL. It also demonstrated rapid bactericidal properties, eliminating pathogens within 30 min, while maintaining low cytotoxicity toward mammalian cells. SAP 2.8 effectively inhibited bacterial biofilm formation and disrupted preformed biofilms. Mechanistic studies revealed that the peptide induces membrane rupture and permeabilization, triggering increase intracellular reactive oxygen species production, ultimately resulting in bacterial death. Notably, SAP 2.8 significantly reduced bacterial load in animal models, positioning it as a promising candidate for the treatment of Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Demeke Asmamaw
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - James Mwangi
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Brenda B. Michira
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Mehwish Khalid
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Prateeksha
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Min Yang
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Qiumin Lu
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Wang Yi
- Center
for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou511458, China
| | - Rebecca Caroline Thuku
- Kunming
Institute of Zoology, University of Chinese
Academy of Sciences, Kunming650223, Yunnan, China
| | - Zilei Duan
- Kunming
Institute of Zoology, Chinese Academy of
Sciences, No.32 Jiaochangdonglu, Kunming650223, YunnanChina
| | - Ren Lai
- Kunming
Institute of Zoology, Chinese Academy of
Sciences, No.32 Jiaochangdonglu, Kunming650223, YunnanChina
| |
Collapse
|
2
|
O'Reilly P, Loiselle G, Darragh R, Slipski C, Bay DC. Reviewing the complexities of bacterial biocide susceptibility and in vitro biocide adaptation methodologies. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:39. [PMID: 40360746 PMCID: PMC12075810 DOI: 10.1038/s44259-025-00108-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025]
Abstract
Decreased bacterial susceptibility to biocides raises concerns due to their influences on antibiotic resistance. The lack of standardized breakpoints, established methods, and consistent terminology complicates this research. This review summarizes techniques for studying biocide resistance mechanisms, susceptibility testing, and in-vitro adaptation methods, highlighting their benefits and limitations. Here, the challenges in studying biocide susceptibility and the need for standardized approaches in biocide research are emphasized for commonly studied biocide classes.
Collapse
Affiliation(s)
- Peter O'Reilly
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Genevieve Loiselle
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ryan Darragh
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Carmine Slipski
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Denice C Bay
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
3
|
Ben-David Y, Sporny M, Brochin Y, Piscon B, Roth S, Zander I, Nisani M, Shoshani S, Yaron O, Karako-Lampert S, Lebenthal-Loinger I, Danielli A, Opatowsky Y, Banin E. SadB, a mediator of AmrZ proteolysis and biofilm development in Pseudomonas aeruginosa. NPJ Biofilms Microbiomes 2025; 11:77. [PMID: 40360526 PMCID: PMC12075610 DOI: 10.1038/s41522-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The ability of bacteria to commit to surface colonization and biofilm formation is a highly regulated process. In this study, we characterized the activity and structure of SadB, initially identified as a key regulator in the transition from reversible to irreversible surface attachment. Our results show that SadB acts as an adaptor protein that tightly regulates the master regulator AmrZ at the post-translational level. SadB directly binds to the C-terminal domain of AmrZ, leading to its rapid degradation, primarily by the Lon protease. Structural analysis suggests that SadB does not directly interact with small molecules upon signal transduction, differing from previous findings in Pseudomonas fluorescens. Instead, the SadB structure supports its role in mediating protein-protein interactions, establishing it as a major checkpoint for biofilm commitment.
Collapse
Affiliation(s)
- Yossi Ben-David
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Michael Sporny
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Yigal Brochin
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Bar Piscon
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Shira Roth
- The Alexander Kofkin Faculty of Engineering, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Itzhak Zander
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Michal Nisani
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Sivan Shoshani
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Orly Yaron
- The Scientific Equipment Center, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Sarit Karako-Lampert
- The Scientific Equipment Center, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Ilana Lebenthal-Loinger
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Amos Danielli
- The Alexander Kofkin Faculty of Engineering, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Ehud Banin
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel.
| |
Collapse
|
4
|
Singh S, Muniz De Oliveira F, Wang C, Kumar M, Xuan Y, DeMazumder D, Sen CK, Roy S. SEMTWIST Quantification of Biofilm Infection in Human Chronic Wound Using Scanning Electron Microscopy and Machine Learning. Adv Wound Care (New Rochelle) 2025. [PMID: 40358506 DOI: 10.1089/wound.2024.0291] [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: 05/15/2025] Open
Abstract
Objective: To develop scanning electron microscopy-based Trainable Weka (Waikato Environment for Knowledge Analysis) Intelligent Segmentation Technology (SEMTWIST), an open-source software tool, for structural detection and rigorous quantification of wound biofilm aggregates in complex human wound tissue matrix. Approach: SEMTWIST model was standardized to quantify biofilm infection (BFI) abundance in 240 distinct SEM images from 60 human chronic wound-edge biospecimens (four technical replicates of each specimen). Results from SEMTWIST were compared against human expert assessments and the gold standard for molecular BFI detection, that is, peptide nucleic acid fluorescence in situ hybridization (PNA-FISH). Results: Correlation and Bland-Altman plot demonstrated a robust correlation (r = 0.82, p < 0.01), with a mean bias of 1.25, and 95% limit of agreement ranging from -43.40 to 47.11, between SEMTWIST result and the average scores assigned by trained human experts. While interexpert variability highlighted potential bias in manual assessments, SEMTWIST provided consistent results. Bacterial culture detected infection but not biofilm aggregates. Whereas the wheat germ agglutinin staining exhibited nonspecific staining of host tissue components and failed to provide a specific identification of BFI. The molecular identification of biofilm aggregates using PNA-FISH was comparable with SEMTWIST, highlighting the robustness of the developed approach. Innovation: This study introduces a novel approach "SEMTWIST" for in-depth analysis and precise differentiation of biofilm aggregates from host tissue elements, enabling accurate quantification of BFI in chronic wound SEM images. Conclusion: Open-source SEMTWIST offers a reliable and robust framework for standardized quantification of BFI burden in human chronic wound-edge tissues, supporting clinical diagnosis and guiding treatment.
Collapse
Affiliation(s)
- Surabhi Singh
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fabio Muniz De Oliveira
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cong Wang
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Manoj Kumar
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yi Xuan
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deeptankar DeMazumder
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chandan K Sen
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sashwati Roy
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Lan J, Zou J, Xin H, Sun J, Han T, Sun M, Niu M. Nanomedicines as disruptors or inhibitors of biofilms: Opportunities in addressing antimicrobial resistance. J Control Release 2025; 381:113589. [PMID: 40032007 DOI: 10.1016/j.jconrel.2025.113589] [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: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
The problem of antimicrobial resistance (AMR) has caused global concern due to its great threat to human health. Evidences are emerging for a critical role of biofilms, one of the natural protective mechanisms developed by bacteria during growth, in resisting commonly used clinical antibiotics. Advances in nanomedicines with tunable physicochemical properties and unique anti-biofilm mechanisms provide opportunities for solving AMR risks more effectively. In this review, we summarize the five "A" stages (adhesion, amplification, alienation, aging and allocation) of biofilm formation and mechanisms through which they protect the internal bacteria. Aimed at the characteristics of biofilms, we emphasize the design "THAT" principles (targeting, hacking, adhering and transport) of nanomedicines in their interactions with biofilms and internal bacteria. Furthermore, recent progresses in multimodal antibacterial nanomedicines, including biofilms disruption and bactericidal activity, and the types of currently available antibiofilm nanomedicines contained organic and inorganic nanomedicines are outlined and highlighted their potential applications in the development of preclinical research. Last but not least, we offer a perspective for the effectiveness of nanomedicines designed to address AMR and challenges associated with their clinical translation.
Collapse
Affiliation(s)
- Jiaming Lan
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jingyu Zou
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - He Xin
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Tao Han
- Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Meng Niu
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
6
|
Verma N, Bajiya M, Dolhey R, Surabhi, Yadav AS, Chaudhary C, Meena D, Arya H, Bhatt TK, Yadav JK, Shukla JN, Swaroop S, Pandey J. Mechanistic Insights into the Antibiofilm Activity of Simvastatin and Lovastatin against Bacillus subtilis. Mol Pharm 2025; 22:2703-2722. [PMID: 40100146 DOI: 10.1021/acs.molpharmaceut.5c00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Statins have been reported for diverse pleiotropic activities, including antimicrobial and antibiofilm. However, due to the limited understanding of their mode of action, none of the statins have gained approval for antimicrobial or antibiofilm applications. In a recent drug repurposing study, we observed that two statins (i.e., Simvastatin and Lovastatin) interact stably with TasA(28-261), the principal extracellular matrix protein of Bacillus subtilis, and also induce inhibition of biofilm formation. Nevertheless, the underlying mechanism remained elusive. In the present study, we examined the impact of these statins on the physiological activity of TasA(28-261), specifically its interaction with TapA(33-253) and aggregation into the amyloid-like structure using purified recombinant TasA(28-261) and TapA(33-253) in amyloid detection-specific in vitro assays (i.e., CR binding and ThT staining assays). Results revealed that both statins interfered with amyloid formation by the TasA(28-261)-TapA(33-253) complex, while neither statin inhibited amyloid formation by lysozyme, a model amyloid-forming protein. Moreover, neither statin significantly altered the expressions of terminal regulatory genes (viz, sinR, sinI) and terminal effector genes (viz, tasA, tapA, and bslA) involved in biofilm formation by B. subtilis. While the intricate interplay between Simvastatin and Lovastatin with the diverse molecular constituents of B. subtilis biofilm remains to be elucidated conclusively, the findings obtained during the present study suggest that the underlying mechanism for Simvastatin- and Lovastatin-mediated inhibition of B. subtilis biofilm formation is manifested by interfering with the aggregation and amyloid formation by TasA(28-261)-TapA(33-253). These results represent one of the first experimental evidence for the underlying mechanism of antibiofilm activity of statins and offer valuable directions for future research to harness statins as antibiofilm therapeutics.
Collapse
Affiliation(s)
- Nidhi Verma
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Mamta Bajiya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Ragini Dolhey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Surabhi
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Abhishek Singh Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Chhavi Chaudhary
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Dhankesh Meena
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Hemant Arya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Tarun K Bhatt
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jay Kant Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jayendra Nath Shukla
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Shiv Swaroop
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Janmejay Pandey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| |
Collapse
|
7
|
Shah T, Zhu C, Shah C, Upadhyaya I, Upadhyay A. Trans-cinnamaldehyde nanoemulsion reduces Salmonella Enteritidis biofilm on steel and plastic surfaces and downregulates expression of biofilm associated genes. Poult Sci 2025; 104:105086. [PMID: 40168703 PMCID: PMC11997393 DOI: 10.1016/j.psj.2025.105086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Salmonella Enteritidis is a major poultry-associated foodborne pathogen that can form sanitizer-tolerant biofilms on various surfaces. The biofilm-forming capability of S. Enteritidis facilitates its survival on farm and food processing equipment. Conventional sanitization methods are not completely effective in killing S. Enteritidis biofilms. This study investigated the efficacy of a Generally Recognized as Safe phytochemical Trans-cinnamaldehyde (TC), and in its nanoemulsion form (TCNE), for inhibiting S. Enteritidis biofilm formation and inactivating mature biofilms developed on polystyrene and stainless-steel surfaces. Moreover, the effect of TC on Salmonella genes critical for biofilm formation was studied. TCNE was prepared using a high energy sonication method with Tween 80. For biofilm inhibition assay, S. Enteritidis was allowed to form biofilms either in the presence or absence of sub-inhibitory concentration (SIC; 0.01 %) of TCNE at 25°C and the biofilm formed was quantified at 24-h intervals for 48 h. For the inactivation assay, S. Enteritidis biofilms developed at 25°C for 48 h were exposed to TCNE (0.5, 1 %) for 1, 5, and 15 min, and surviving S. Enteritidis in the biofilm were enumerated. SIC of TCNE inhibited S. Enteritidis biofilm by 45 % on polystyrene and 75 % on steel surface after 48 h at 25°C compared to control (P < 0.05). All TCNE treatments rapidly inactivated S. Enteritidis mature biofilm on polystyrene and steel surfaces (P < 0.05). The lower concentration of TCNE (0.5 %) reduced S. Enteritidis counts by 1.5 log CFU/ml as early as 1 min of exposure on both polystyrene and stainless-steel surfaces. After 15 min of exposure, TCNE at concentration of 0.5 or 1 % reduced S. Enteritidis count significantly by 4.5 log CFU or 6 log CFU/ml on polystyrene or stainless-steel surfaces. TC downregulated the expression of S. Enteritidis genes (hilA, hilC, flhD, csgA, csgD, sdiA) responsible for biofilm formation (P < 0.05). Results suggest that TCNE has potential as a natural disinfectant for controlling S. Enteritidis biofilms on common farm and food processing surfaces, such as plastic and steel.
Collapse
Affiliation(s)
- Trushenkumar Shah
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Chen Zhu
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Chetna Shah
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Indu Upadhyaya
- Department of Extension, University of Connecticut, Storrs, Connecticut, USA
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA.
| |
Collapse
|
8
|
Fares M, Imberty A, Titz A. Bacterial lectins: multifunctional tools in pathogenesis and possible drug targets. Trends Microbiol 2025:S0966-842X(25)00083-6. [PMID: 40307096 DOI: 10.1016/j.tim.2025.03.007] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 05/02/2025]
Abstract
Glycans are vital macromolecules with diverse biological roles, decoded by lectins - specialized carbohydrate-binding proteins crucial in pathogenesis. The WHO identifies bacterial antimicrobial resistance (AMR) as a critical global health challenge, necessitating innovative strategies that also target non-antibiotic pathways. Recent studies highlight bacterial lectins as key players in pathogenesis and promising therapeutic targets, with early clinical success using glycomimetics and vaccines to treat and prevent AMR-related infections. This review covers the current knowledge on bacterial lectins, their classifications, and roles in host recognition and adhesion, biofilm formation, cytotoxicity, and host immune evasion, with examples of well-characterized lectins. It also explores their therapeutic potential and highlights novel lectins with unknown functions, encouraging further research.
Collapse
Affiliation(s)
- Mario Fares
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, D-66123 Saarbrücken, Germany
| | - Anne Imberty
- University Grenoble Alpes, CNRS, CERMAV, 601 rue de la chimie, Grenoble 38000, France
| | - Alexander Titz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, D-66123 Saarbrücken, Germany.
| |
Collapse
|
9
|
Bull F, Tavaddod S, Bommer N, Perry M, Brackley CA, Allen RJ. Different factors control long-term versus short-term outcomes for bacterial colonisation of a urinary catheter. Nat Commun 2025; 16:3940. [PMID: 40287414 PMCID: PMC12033313 DOI: 10.1038/s41467-025-59161-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Urinary catheters are used extensively in hospitals and long-term care and they are highly prone to infection. Understanding the pathways by which bacteria colonise a urinary catheter could guide strategies to mitigate infection, but quantitative models for this colonisation process are lacking. Here we present a mathematical model for bacterial colonisation of a urinary catheter that integrates population dynamics and fluid dynamics. The model describes bacteria migrating up the outside surface of the catheter, spreading into the bladder and being swept through the catheter lumen. Computer simulations of the model reveal that clinical outcomes for long-term versus short-term catheterisation are controlled by different factors: the rate of urine production by the kidneys as opposed to urethral length, catheter surface properties and bacterial motility. Our work may help explain variable susceptibility to catheter-associated urinary tract infection (CAUTI) among individuals and the mixed success of antimicrobial surface coatings. Our model suggests that for long-term catheterised patients, increasing fluid intake or reducing residual urine volume in the bladder may help prevent infection, while antimicrobial surface coatings are predicted to be effective only for short-term catheterised patients. Therefore, different catheter management strategies could be rationally targeted to long-term vs short-term catheterised patients.
Collapse
Affiliation(s)
- Freya Bull
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
- Department of Mathematics, University College London, London, UK
| | - Sharareh Tavaddod
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Nick Bommer
- Veterinary Specialists Scotland, Livingston, UK
| | - Meghan Perry
- Clinical Infection Research Group, Regional Infectious Diseases Unit, Western General Hospital, Edinburgh, UK
| | - Chris A Brackley
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Rosalind J Allen
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK.
- Theoretical Microbial Ecology, Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
10
|
Hung CH, Chan KH, Kong WP, Du RL, Ding K, Liang Z, Wang Y, Wong KY. A Water-Soluble Aggregation-Induced Emission Photosensitizer with Intrinsic Antibacterial Activity as an Antiplanktonic and Antibiofilm Therapeutic Agent. J Med Chem 2025; 68:8768-8785. [PMID: 40186565 PMCID: PMC12035805 DOI: 10.1021/acs.jmedchem.5c00403] [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: 02/10/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Photosensitizers (PSs) with aggregation-induced emission (AIE) properties have gained popularity for treating bacterial infections. However, most AIE PSs have a poor water solubility and low selectivity, limiting their applications in biological systems. Herein, we report a water-soluble and bacteria-targeting AIE PS that exhibits minimum cytotoxicity toward human cells with and without light irradiation. Acting as a narrow-spectrum antibacterial agent without light irradiation, TPA-1 eradicates planktonic Staphylococcus aureus and inhibits biofilm formation by targeting the S. aureus membrane, inhibiting the supercoiling activity of S. aureus DNA gyrase, and causing the downregulation of multiple essential proteins. Upon light irradiation, TPA-1 generates reactive oxygen species (ROS) that cause membrane damage, resulting in excellent antiplanktonic and antibiofilm activities against S. aureus and Pseudomonas aeruginosa, significantly reducing the number of viable bacteria in biofilms and promoting wound healing in vivo.
Collapse
Affiliation(s)
- Cheung-Hin Hung
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Ka Hin Chan
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Wai-Po Kong
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Ruo-Lan Du
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Kang Ding
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Zhiguang Liang
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Yong Wang
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical
Biology and Drug Discovery, Department of Applied Biology and Chemical
Technology, The Hong Kong Polytechnic University, Kowloon, Hong
Kong, China
| |
Collapse
|
11
|
Shahrour H, Ferreira DA, Sheridan L, Fitzgerald-Hughes D, O’Gara JP, Devocelle M, Kelly H, O’Neill E. Potent antimicrobial activity of hydrogel loaded with the antimicrobial peptide, D-Bac8c 2,5 Leu, against monospecies and polymicrobial biofilms of Staphylococcus aureus and Pseudomonas aeruginosa. Front Microbiol 2025; 16:1571649. [PMID: 40342603 PMCID: PMC12058896 DOI: 10.3389/fmicb.2025.1571649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Introduction Acute and chronic wound infections involving biofilms and caused by antimicrobial resistant (AMR) pathogens present significant challenges in healthcare, leading to substantial patient morbidity, increased hospital stays, and rising healthcare costs. Novel antimicrobial therapies are urgently needed to address these infections. Methods A screening of multiple antimicrobial peptides (AMPs) was performed and the most potent candidate, D-Bac8c2,5 Leu, was tested against monospecies and polymicrobial biofilms of Staphylococcus aureus and Pseudomonas aeruginosa using static and dynamic in vitro models. Cytotoxicity was evaluated on human cell lines, and the peptide was incorporated into a methylcellulose hydrogel to assess sustained release and antimicrobial efficacy as a hydrogel dressing. Results D-Bac8c2,5 Leu significantly reduced biofilm viability in both monospecies and polymicrobial biofilms. In static biofilm assays, treatment led to a 2-3 log reduction in bacterial load compared to untreated controls. In Duckworth biofilm flow device, a similar reduction was observed, demonstrating efficacy in conditions mimicking wound environments. Furthermore, D-Bac8c2,5 Leu exhibited low cytotoxicity against human cell lines, and its incorporation into a methylcellulose hydrogel facilitated sustained release and enhanced antimicrobial activity. Furthermore, the peptide-loaded hydrogel showed considerable efficacy in disrupting pre-formed biofilms, underscoring its potential as a novel treatment for acute and chronic wound infections. Discussion These findings highlight the potential of D-Bac8c2,5 Leu to help address the urgent need for effective therapies against AMR pathogens and biofilm-associated wound infections. Further studies should focus on in vivo efficacy to optimize its therapeutic application in wound care.
Collapse
Affiliation(s)
- Hawraa Shahrour
- Department of Clinical Microbiology, RCSI Education and Research Centre, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Daniela Alves Ferreira
- Department of Clinical Microbiology, RCSI Education and Research Centre, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luke Sheridan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Deirdre Fitzgerald-Hughes
- Department of Clinical Microbiology, RCSI Education and Research Centre, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - James P. O’Gara
- Department of Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Marc Devocelle
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Helena Kelly
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Eoghan O’Neill
- Department of Clinical Microbiology, RCSI Education and Research Centre, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Microbiology, Connolly Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Sajeevan A, Andrew DJ, Patra TN, Solomon AP, Dandela R. Novel synthesis and anti-pathogenic properties of ensifentrine and its intermediates against Pseudomonas aeruginosa. RSC Adv 2025; 15:13053-13063. [PMID: 40271403 PMCID: PMC12015936 DOI: 10.1039/d5ra01722j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive respiratory disorder marked by persistent lung inflammation and airway constriction. It presents a formidable global health challenge owing to its high morbidity and mortality rates. It is often aggravated by infections from pathogens such as Pseudomonas aeruginosa, a predominant pathogen that accelerates lung function deterioration and triggers frequent exacerbations. Ensifentrine (ENF) exhibits strong anti-inflammatory effects and is a selective dual inhibitor of the enzymes PDE3 and PDE4, which have been reported to be beneficial in treating COPD exacerbation. This study examined the anti-pathogenic activity of ENF against P. aeruginosa by adopting an innovative synthetic route. A series of intermediates were synthesized via the novel route, optimizing the yield and integrity of ENF. Further investigations to determine the activity of the compound against P. aeruginosa involved antibacterial and antibiofilm testing and identification of the potential mechanisms of action. Preliminary results demonstrate that ENF and its intermediate ENFA exhibit 50-60% robust biofilm-inhibition and biofilm-eradication effects at remarkably low concentrations of 3.9 μM and 7.9 μM, respectively. Furthermore, ENF disrupts quorum sensing, leading to a 35% reduction in the production of pyoverdine and exopolysaccharide, which are two key virulence factors of P. aeruginosa. Importantly, ENF exhibits synergistic activity with ciprofloxacin, further enhancing its antimicrobial efficacy at a concentration of 0.25 μg mL-1. This study focuses on the innovative synthesis of ENF and its promising anti-pathogenic properties, which may make it an effective adjunctive treatment for COPD caused by P. aeruginosa.
Collapse
Affiliation(s)
- Anusree Sajeevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur India
| | - Deepthi Joseph Andrew
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur India
| | - T Nalinikanta Patra
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology Bhubaneswar Odisha India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University Thanjavur India
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology Bhubaneswar Odisha India
| |
Collapse
|
13
|
Cucu CI, Giurcăneanu C, Poenaru E, Popa LG, Popa MI, Chifiriuc MC, Lazăr V, Holban AM, Gheorghe-Barbu I, Muntean AA, Caracoti CȘ, Mihai MM. Phenotypic and Genotypic Bacterial Virulence and Resistance Profiles in Hidradenitis Suppurativa. Int J Mol Sci 2025; 26:3502. [PMID: 40331997 PMCID: PMC12026681 DOI: 10.3390/ijms26083502] [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: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic inflammatory skin condition, primarily affecting young individuals, with a significant impact on their quality of life due to recurrent, painful nodules, abscesses, and oozing sinus tracts, primarily affecting intertriginous areas. The pathogenesis of HS is multifactorial, involving a complex interplay between genetic predisposition, immune dysregulation, microbial, and environmental factors. While it is known that cutaneous and gut microbiome contribute to innate immune dysregulation in HS, their precise involvement in disease pathogenesis remains unclear. Despite several studies investigating the microbiome of HS lesions, either by culture-dependent or independent methods, there is no data available on the interplay between bacterial virulence profiles, clinical manifestations, and the host immune response. This study aimed to explore the phenotypic and genotypic resistance and virulence profiles of microorganisms isolated from HS lesions (including the expression of soluble virulence factors and the ability to develop biofilms), with a special focus on Staphylococcus aureus (S. aureus), one of the most frequent infectious agents of HS. A total of 92 bacterial strains, belonging to 20 different bacterial species, were isolated from the HS lesions of 23 patients. The strains of Staphylococcus, Corynebacterium, and Enterococcus expressed the highest levels of soluble virulence factors, such as hemolysins, lecithinase, and lipase, which are involved in bacterial persistence, local invasivity, and tissue damage. Moreover, a significant variation among bacterial species was noted regarding the capacity to develop biofilms, with a potential impact on disease chronicization, bacterial tolerance to antibiotics, and immune defense mechanisms. The genetic characterization of methicillin-resistant staphylococci revealed the presence of adhesins, hemolysin and enterotoxin genes as well as methicillin and macrolides resistance genes. Our findings highlight the critical role of virulence determinants, including bacterial biofilms, in HS pathogenesis, emphasizing the need for targeted therapeutic strategies to disrupt biofilms and mitigate infection severity.
Collapse
Affiliation(s)
- Corina Ioana Cucu
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (L.G.P.); (M.M.M.)
| | - Călin Giurcăneanu
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (L.G.P.); (M.M.M.)
| | - Elena Poenaru
- Department of Medical Informatics and Biostatistics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (L.G.P.); (M.M.M.)
| | - Mircea Ioan Popa
- Department of Microbiology II, “Cantacuzino” Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.I.P.); (C.Ș.C.)
| | - Mariana Carmen Chifiriuc
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050663 Bucharest, Romania (V.L.); (I.G.-B.)
| | - Veronica Lazăr
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050663 Bucharest, Romania (V.L.); (I.G.-B.)
| | - Alina Maria Holban
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050663 Bucharest, Romania (V.L.); (I.G.-B.)
| | - Irina Gheorghe-Barbu
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050663 Bucharest, Romania (V.L.); (I.G.-B.)
| | - Andrei-Alexandru Muntean
- Department of Microbiology II, “Cantacuzino” Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.I.P.); (C.Ș.C.)
| | - Costin Ștefan Caracoti
- Department of Microbiology II, “Cantacuzino” Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.I.P.); (C.Ș.C.)
| | - Mara Mădălina Mihai
- Department of Oncologic Dermatology, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (L.G.P.); (M.M.M.)
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050663 Bucharest, Romania (V.L.); (I.G.-B.)
| |
Collapse
|
14
|
Yao XF, Wang YJ, Lin YS. Palliative care with negative pressure wound therapy application in malignant wounds: a systematic review. J Wound Care 2025; 34:304-311. [PMID: 40227927 DOI: 10.12968/jowc.2021.0254] [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: 04/16/2025]
Abstract
OBJECTIVE To synthesise and appraise the evidence for and benefits of palliative application of negative pressure wound therapy (NPWT) in malignant wounds. METHOD We performed a systematic review according to Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA). MEDLINE, Embase, Cochrane Library and trial registers (www.clinicaltrials.gov) from inception to 1 June 2021 were searched. Quality was assessed using the tool of methodological quality and synthesis from the 2018 BMJ Evidence-Based Medicine article on case series and case report synthesis. RESULTS Of 765 articles screened, 14 eligible studies were included in the review. All were case reports and included a total of 22 patients. The mean age of the patients was 60.7 (range: 33-77) years. The location of the hard-to-heal wounds was widely distributed: five wounds on the scalp; three wounds over the anogenital area; and the remaining wounds on the trunk and extremities. The reported outcomes included: improvement of wound healing; decreased exudation and malodour of the wound; decreased pain sensation; eligibility for further treatment; and a shift to the homecare system. CONCLUSION The findings of this study suggested NPWT could be a choice in palliative care for patients with malignant wounds. However, more studies are needed to evaluate the efficacy of NPWT in these wounds.
Collapse
Affiliation(s)
- Xiao-Feng Yao
- MD, Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yen-Jen Wang
- MD, Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan
- MD, Department of Cosmetic Applications and Management, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Yang-Sheng Lin
- MD, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- MD, Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- MD, Evidence-Based Medicine Center, MacKay Memorial Hospital, Taipei, Taiwan
- MD, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Varin-Simon J, Haney EF, Colin M, Velard F, Gangloff SC, Hancock REW, Reffuveille F. D-enantiomeric antibiofilm peptides effective against anaerobic Cutibacterium acnes biofilm. Microbiol Spectr 2025; 13:e0252324. [PMID: 40130849 PMCID: PMC12053997 DOI: 10.1128/spectrum.02523-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/27/2025] [Indexed: 03/26/2025] Open
Abstract
The emergence of antibiotic resistance, biofilm formation, and internalization by host cells contribute to a high risk of chronic infections, highlighting the necessity to develop novel therapeutic strategies. Identification of natural host defense peptides (HDPs) with promising antimicrobial and antibiofilm activities led to the development of synthetic peptides with broad-spectrum efficacy. However, few studies have examined their effect on anaerobic bacterial species. This study aimed to test the effect of synthetic HDPs on Cutibacterium acnes, an anaerobe species involved in 10% of prosthesis joint infections (PJI). A preliminary screen identified three peptides (DJK5, AB009-D, and AB101-D) with promising activity against four C. acnes strains (two of which were isolated from PJI). A bactericidal effect was observed for the three peptides with 50% of planktonic bacteria killing for AB009-D and AB101-D after only 3 hours of contact. DJK5 and AB009-D inhibited the C. acnes adhesion on plastic and titanium supports with a 2-log decrease in bacterial cells. In the presence of peptides, the morphology of C. acnes cells was altered with an increase in cell length observed, especially for one of the non-PJI-related strains. Against mature biofilms, AB101-D was the most effective with an approximate 2-log decrease in adhered CFUs, indicating the induction of bacterial dispersion or death. DJK5 also inhibited C. acnes internalization by osteoblasts, with a reduction of the internalized bacteria quantity for three strains. Overall, this study demonstrates that synthetic HDPs are effective against anaerobic bacteria and hold promise as novel therapeutic candidates to prevent or treat C. acnes PJIs.IMPORTANCEThe emergence of antibiotic tolerance highlights the necessity to develop novel therapeutic strategies with promising antimicrobial but also antibiofilm activities. In this study, we tested the effect of synthetic host defense peptides (HDPs) on Cutibacterium acnes, an anaerobic species, rarely studied, whereas involved in 10% of prosthesis joint infections (PJI). In our study, we demonstrate that the selected synthetic HDPs are effective against this anaerobic bacteria, both as a preventive treatment (effect on planktonic growth, bacterial adhesion, and biofilm formation) and against internalization of C. acnes by osteoblasts, revealing that these peptides are promising as novel therapeutic candidates to prevent or treat C. acnes PJIs.
Collapse
Affiliation(s)
- Jennifer Varin-Simon
- Université de Reims Champagne-Ardenne, BIOS, Reims, Grand Est, France
- Université de Reims Champagne-Ardenne, UFR Pharmacie, Reims, Grand Est, France
| | - Evan F. Haney
- Asep Medical Inc./ABT Innovations Inc., Victoria, British Columbia, Canada
- Department of Microbiology and Immunology, Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marius Colin
- Université de Reims Champagne-Ardenne, BIOS, Reims, Grand Est, France
- Université de Reims Champagne-Ardenne, UFR Pharmacie, Reims, Grand Est, France
| | - Frédéric Velard
- Université de Reims Champagne-Ardenne, BIOS, Reims, Grand Est, France
| | - Sophie C. Gangloff
- Université de Reims Champagne-Ardenne, BIOS, Reims, Grand Est, France
- Université de Reims Champagne-Ardenne, UFR Pharmacie, Reims, Grand Est, France
| | - Robert E. W. Hancock
- Asep Medical Inc./ABT Innovations Inc., Victoria, British Columbia, Canada
- Department of Microbiology and Immunology, Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fany Reffuveille
- Université de Reims Champagne-Ardenne, BIOS, Reims, Grand Est, France
- Université de Reims Champagne-Ardenne, UFR Pharmacie, Reims, Grand Est, France
| |
Collapse
|
16
|
Sakib S, Andoy NMO, Yang JYC, Galang A, Sullan RMA, Zou S. Antimicrobial and anti-inflammatory effects of polyethyleneimine-modified polydopamine nanoparticles on a burn-injured skin model. Biomater Sci 2025; 13:1770-1783. [PMID: 39995391 DOI: 10.1039/d4bm01530d] [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: 02/26/2025]
Abstract
Chronic infections involving bacterial biofilms pose significant treatment challenges due to the resilience of biofilms against existing antimicrobials. Here, we introduce a nanomaterial-based platform for treating Staphylococcus epidermidis biofilms, both in isolation and within a biofilm-infected burn skin model. Our approach leverages biocompatible and photothermal polydopamine nanoparticles (PDNP), functionalized with branched polyethyleneimine (PEI) and loaded with the antibiotic rifampicin, to target bacteria dwelling within biofilms. A key innovation of our method is its ability to not only target planktonic S. epidermidis but also effectively tackle biofilm-embedded bacteria. We demonstrated that PDNP-PEI interacts effectively with the bacterial surface, facilitating laser-activated photothermal eradication of planktonic S. epidermidis. In a 3D skin burn injury model, PDNP-PEI demonstrates anti-inflammatory and reactive oxygen species (ROS)-scavenging effects, reducing inflammatory cytokine levels and promoting healing. The rifampicin-loaded PDNP-PEI (PDNP-PEI-Rif) platform further shows significant efficacy against bacteria inside biofilms. The PDNP-PEI-Rif retained its immunomodulatory activity and efficiently eradicated biofilms grown on our burn-injured 3D skin model, effectively addressing the challenges of biofilm-related infections. This achievement marks a significant advancement in infection management, with the potential for a transformative impact on clinical practice.
Collapse
Affiliation(s)
- Sadman Sakib
- Metrology Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1N 5A2, Canada.
| | - Nesha May O Andoy
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
| | - Jessica Y C Yang
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
| | - Anna Galang
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON, M5S 3H6, Canada.
| | - Ruby May A Sullan
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON, M5S 3H6, Canada.
| | - Shan Zou
- Metrology Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1N 5A2, Canada.
| |
Collapse
|
17
|
Rubio C, Ramírez J, Weinstein-Oppenheimer C, Bahamondez-Canas TF, Quiñones N. Isolation, Chemical Characterization, and Antimicrobial Activity of Secondary Metabolites from Pseudocyphellaria faveolata. Molecules 2025; 30:1368. [PMID: 40142140 PMCID: PMC11944333 DOI: 10.3390/molecules30061368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
INTRODUCTION Antimicrobial resistance is a global threat, highlighting the urgent need for novel antimicrobial agents. Among the mechanisms of resistance, bacteria can release drug-degrading enzymes and express efflux pumps, as well as grow in protected aggregates known as biofilms. Pseudomonas aeruginosa and Staphylococcus aureus are among the most prevalent biofilm infections in chronic wounds, respiratory and urinary tract infections, and device-associated infections. Pseudocyphellaria faveolata (Delise) Malme is a lichen with metabolites with unexplored antimicrobial potential. AIMS To identify and characterize the major metabolites present in Pseudocyphellaria Faveolata and to determine their antimicrobial activity against Staphylococcus aureus and Pseudomonas aeruginosa. METHODS The molecules were purified by column chromatography and characterized by NMR spectroscopy. The antimicrobial activity of the compounds was determined in terms of proliferation, adhesion, and viability against P. aeruginosa and S. aureus by the broth microdilution method and crystal violet staining. Viability was determined by the resazurin reduction assay on normal human fibroblasts to determine cytotoxicity over human cells. RESULTS The major metabolites were spectroscopically characterized and identified as physciosporin and methyl virensate. Physciosporin showed antimicrobial activity on S. aureus, with a MIC of 32 μg/mL and MBC of 128 μg/mL, and prevented biofilm formation from 16 μg/mL. Methyl virensate also had antimicrobial activity on S. aureus (MIC = 64 μg/mL). None of these metabolites significantly affected P. aeruginosa proliferation, viability, or adhesion. Cytotoxicity of physciosporin at 16 ug/mL on normal human fibroblasts was below 20%. CONCLUSIONS This is the first report on the study of the antimicrobial activity of these compounds. Physciosporin showed promising activity in preventing the formation of S. aureus biofilms, which are responsible for chronic infections. These findings provide a foundation for exploring the antimicrobial potential of other lichenic depsidones.
Collapse
Affiliation(s)
- Cecilia Rubio
- Herbario de Líquenes, Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile; cecilia.rubio-@uv.cl (C.R.); (J.R.)
- Magíster en Gestión Farmacéutica y Farmacia Asistencial, Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Javiera Ramírez
- Herbario de Líquenes, Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile; cecilia.rubio-@uv.cl (C.R.); (J.R.)
| | - Caroline Weinstein-Oppenheimer
- Laboratorio de Innovación Terapéutica y Diagnóstico Molecular, Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile;
- Centro de Investigación, Desarrollo e Innovación de Productos Bioactivos (CInBIO), Universidad de Valparaíso, Valparaíso 2340000, Chile;
| | - Tania F. Bahamondez-Canas
- Centro de Investigación, Desarrollo e Innovación de Productos Bioactivos (CInBIO), Universidad de Valparaíso, Valparaíso 2340000, Chile;
- Laboratorio de Farmacotecnia Antimicrobiana (LADEFAM), Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Natalia Quiñones
- Herbario de Líquenes, Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile; cecilia.rubio-@uv.cl (C.R.); (J.R.)
- Centro de Investigación, Desarrollo e Innovación de Productos Bioactivos (CInBIO), Universidad de Valparaíso, Valparaíso 2340000, Chile;
| |
Collapse
|
18
|
Wang W, Zhong Q, Huang X. Antibacterial and anti-biofilm activities of Derazantinib (ARQ-087) against Staphylococcus aureus. Arch Microbiol 2025; 207:78. [PMID: 40047947 DOI: 10.1007/s00203-025-04288-1] [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: 01/24/2025] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
The global rise of multidrug-resistant pathogens, particularly methicillin-resistant Staphylococcus aureus (MRSA), represents a critical public health challenge. This study evaluates the antibacterial and anti-biofilm activities of Derazantinib (ARQ-087) against S. aureus. ARQ-087 exhibited minimum inhibitory concentration (MIC) values ranging from 4 to 16 µM against S. aureus reference laboratory strains and diverse clinical MRSA isolates, demonstrating strong antibacterial activity with minimal resistance development. Time-kill assays demonstrated a concentration- and time-dependent reduction in bacterial viability. Crystal violet staining assays revealed that ARQ-087 significantly inhibited MRSA biofilm formation in a dose-dependent manner. Additionally, ARQ-087 exhibited strong anti-biofilm activity against pre-formed biofilms, as shown by colony counts and confocal laser scanning microscopy, which indicated extensive biofilm disruption and bacterial cell death. Mechanistic studies revealed that ARQ-087 disrupts bacterial membrane integrity, as evidenced by SYTOX Green and DISC3(5) fluorescence assays, while inducing intracellular ATP depletion and reactive oxygen species generation, contributing to bacterial death. ARQ-087 also displayed negligible hemolytic activity and no acute toxicity observed in a Galleria mellonella infection model. In this model, ARQ-087 prolonged the survival of larvae infected with S. aureus. These findings highlight ARQ-087 as a promising therapeutic candidate for treating MRSA infections and biofilm-associated diseases. Further preclinical studies are needed to confirm its potential for clinical application.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Clinical Laboratory, The First Hospital of Nanchang, Nanchang, Jiangxi, People's Republic of China
| | - Qiuxiang Zhong
- Department of Clinical Laboratory, The First Hospital of Nanchang, Nanchang, Jiangxi, People's Republic of China
| | - Xincheng Huang
- Department of Clinical Laboratory, The First Hospital of Nanchang, Nanchang, Jiangxi, People's Republic of China.
| |
Collapse
|
19
|
Gondil V, Ashcraft M, Ghalei S, Kumar A, Wilson SN, Devine R, Handa H, Brisbois EJ. Anti-Infective Bacteriophage Immobilized Nitric Oxide-Releasing Surface for Prevention of Thrombosis and Device-Associated Infections. ACS APPLIED BIO MATERIALS 2025; 8:1362-1376. [PMID: 39895136 PMCID: PMC11836933 DOI: 10.1021/acsabm.4c01638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/31/2024] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
The treatment of critically ill patients has made great strides in the past few decades due to the rapid development of indwelling medical devices. Despite immense advancements in the design of these devices, indwelling medical device-associated infections and thrombosis are two major clinical problems that may lead to device failure and compromise clinical outcomes. Antibiotics are the current treatment choice for these infections; however, the global emergence of antibiotic-resistance and their biofilm formation abilities complicate the management of such infections. Moreover, systemic administration of anticoagulants has been used to counter medical device-induced thrombosis, but a range of serious adverse effects associated with all types of available anticoagulants entails exploring alternative options to counter device-associated thrombosis. In this study, bacteriophages (phages) were covalently immobilized on polydimethylsiloxane (PDMS) surface containing the nitric oxide (NO) donor S-nitroso-N-acetylpenicillamine (SNAP) via SNAP impregnation method. This dual strategy combines the targeted antibacterial activity of phages against bacterial pathogens with the antibacterial-antithrombotic activity of NO released from the polymeric surface. The PDMS, SNAP-PDMS, phage-immobilized PDMS (PDMS-Phage), and phage-immobilized SNAP-PDMS (SNAP-PDMS-Phage) surfaces were characterized for their surface topology, elemental composition, contact angle, SNAP loading, NO release and phage distribution. SNAP-PDMS and SNAP-PDMS-Phage surfaces showed similar and consistent NO release profiles over 24 h of incubation. Immobilization of whole phages on PDMS and SNAP-PDMS was achieved with densities of 2.4 ± 0.54 and 2.1 ± 0.33 phages μm-2, respectively. Immobilized phages were found to retain their activity, and SNAP-PDMS-Phage surfaces showed a significant reduction in planktonic (99.99 ± 0.08%) as well as adhered (99.80 ± 0.05%) Escherichia coli as compared to controls in log killing assays. The SNAP-PDMS-Phage surfaces also exhibited significantly reduced platelet adhesion by 64.65 ± 2.95% as compared to control PDMS surfaces. All fabricated surfaces were found to be nonhemolytic and do not exhibit any significant cytotoxic effects toward mammalian fibroblast cells. This study is the first of its kind to demonstrate the combinatorial pertinence of phages and NO to prevent antibiotic-resistant/sensitive bacterial infections and thrombosis associated with indwelling medical devices.
Collapse
Affiliation(s)
- Vijay
Singh Gondil
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Morgan Ashcraft
- Pharmaceutical
and Biomedical Sciences Department, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Sama Ghalei
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Anil Kumar
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Sarah N. Wilson
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Ryan Devine
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Hitesh Handa
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
- Pharmaceutical
and Biomedical Sciences Department, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Elizabeth J. Brisbois
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
20
|
Nagoba BS, Rayate AS, Gavkare AM, Rao A. The efficacy and safety of acids as topical antimicrobial agents: a review. J Wound Care 2025; 34:119-127. [PMID: 39928472 DOI: 10.12968/jowc.2023.0176] [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: 02/12/2025]
Abstract
OBJECTIVE Infected wounds, refractory to conventional therapy, are a major burden on the healthcare system. Available data show that many commonly used antiseptic agents may be toxic to the cells involved in the healing process and may affect the normal tissue repair. The topical use of different acids to control wound infections effectively and promote healing is well known. The present review aims to summarise the safety and efficacy of various acids as topical agents for treating wound infections. METHOD A literature search was performed in PubMed and manually from other sources (cross references and journal sites). RESULTS We reviewed 116 articles, from which data from 86 relevant articles were analysed. The studies showed that various organic acids were clinically effective in treating wound infections. CONCLUSION This study found that various organic acids can act as a substitute for antiseptics to control wound infections refractory to conventional antibiotic therapy and local wound care. Various organic acids differ in efficacy, safety and limitations as topical agents to control wound infections and promote healing. Some acids deliver better results than others, particularly in those cases in which antibiotics and routine antiseptic agents yield little lasting success, especially in controlling hospital strains with multiple antibiotic resistance. Among topically used acids, citric acid and acetic acid are associated with better results.
Collapse
Affiliation(s)
| | | | - Ajay M Gavkare
- Department of Physiology, MIMSR Medical College, Latur, India
| | - Arunkumar Rao
- Department of Orthopaedics, MIMSR Medical College, Latur, India
| |
Collapse
|
21
|
Fakher S, Westenberg D. Evaluation of the antibacterial properties of four bioactive biomaterials for chronic wound management. Future Microbiol 2025; 20:247-258. [PMID: 39810612 PMCID: PMC11812403 DOI: 10.1080/17460913.2025.2453334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025] Open
Abstract
AIM Chronic wound infections present a prevalent medical issue and a multifaceted problem that significantly impacts healthcare systems worldwide. Biofilms formed by pathogenic bacteria are fundamental virulence factors implicated in the complexity and persistence of bacterial-associated wound infections, leading to prolonged recovery times and increased risk of infection. This study aims to investigate the antibacterial effectiveness of commonly employed bioactive wound healing compositions with a particular emphasis on their effectiveness against common bacterial pathogens encountered in chronic wounds - Staphylococcus epidermidis, Escherichia coli, and Pseudomonas aeruginosa to identify optimal wound product composition for managing chronic wound infections. METHODS This study tested the antibacterial and antibiofilm effectiveness of four bioactive wound healing materials by performing in vitro antibacterial assays and measuring ion release profiles. RESULTS The anti-biofilm effectiveness differed extensively among the biomaterials tested and slightly among the bacterial species. Particularly, copper and zinc-doped borate bioactive glass wound healing compositions inhibited the three clinically relevant bacteria in both planktonic and biofilm forms, which were found to be ascribed to the copper and zinc gradual release. CONCLUSION The findings suggest that copper and zinc-doped bioactive glasses hold great promise for improving chronic wound management by providing strong antibacterial action and promoting faster healing.
Collapse
Affiliation(s)
- Sarah Fakher
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - David Westenberg
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| |
Collapse
|
22
|
Rabbitt D, Villapún VM, Carter LN, Man K, Lowther M, O'Kelly P, Knowles AJ, Mottura A, Tang YT, Luerti L, Reed RC, Cox SC. Rethinking Biomedical Titanium Alloy Design: A Review of Challenges from Biological and Manufacturing Perspectives. Adv Healthc Mater 2025; 14:e2403129. [PMID: 39711273 PMCID: PMC11804846 DOI: 10.1002/adhm.202403129] [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: 08/20/2024] [Revised: 11/14/2024] [Indexed: 12/24/2024]
Abstract
Current biomedical titanium alloys have been repurposed from other industries, which has contributed to several biologically driven implant failure mechanisms. This review highlights the added value that may be gained by building an appreciation of implant biological responses at the onset of alloy design. Specifically, the fundamental mechanisms associated with immune response, angiogenesis, osseointegration and the potential threat of infection are discussed, including how elemental selection can modulate these pivotal systems. With a view to expedite inclusion of these interactions in alloy design criteria, methods to analyze these performance characteristics are also summarized. While machine learning techniques are being increasingly used to unearth complex relationships between alloying elements and material properties, much is still unknown about the correlation between composition and some bio-related properties. To bridge this gap, high-throughput methods are also reviewed to validate biological response along with cutting edge manufacturing approaches that may support rapid discovery. Taken together, this review encourages the alloy development community to rethink their approach to enable a new generation of biomedical implants intrinsically designed for a life in the body, including functionality to tackle biological challenges thereby offering improved patient outcomes.
Collapse
Affiliation(s)
- Daisy Rabbitt
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Victor M. Villapún
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Luke N. Carter
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Kenny Man
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht3584 CTThe Netherlands
| | - Morgan Lowther
- Paihau‐Robinson Research InstituteVictoria University of WellingtonWellington5010New Zealand
| | - Paraic O'Kelly
- Center for the Accelerated Maturation of MaterialsDepartment of Materials Science and EngineeringThe Ohio State University1305 Kinnear RoadColumbusOH43212USA
| | | | - Alessandro Mottura
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
| | - Yuanbo T. Tang
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
| | - Lorenzo Luerti
- Alloyed LtdUnit 15, Oxford Industrial ParkYarntonOX5 1QUUK
| | - Roger C. Reed
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
- Department of MaterialsUniversity of OxfordParks RoadOxfordOX1 3PJUK
| | - Sophie C. Cox
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| |
Collapse
|
23
|
Amer AM, Charnock C, Nguyen S. The impact of surface charge on the interaction of cholesterol-free fusogenic liposomes with planktonic microbial cells and biofilms. Int J Pharm 2025; 669:125088. [PMID: 39706381 DOI: 10.1016/j.ijpharm.2024.125088] [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: 10/24/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
This study focused on the development of cholesterol-free fusogenic liposomes with different surface charge with the aim of improving biofilm penetration. In vitro assessments of the liposomes included physical stability, biocompatibility, fusion with microbial cells, and the ability to penetrate established biofilms. Using dynamic light scattering, cholesterol-free, fusogenic liposomes were found to be < 200 nm in size with small size distribution (PDI < 0.1) and physically stable for a year when stored at 4 °C. Transmission electron microscopy (TEM) images confirmed vesicular sizes for selected liposomal formulations. Liposomal ability to fuse with microbial cells was assessed using lipid mixing and flow cytometer assays. Fusion levels were found to be higher with Escherichia coli compared to Staphylococcus aureus and Candida albicans regardless of the liposomal charge. Neutral liposomes exhibited highest fusion, followed by cationic and anionic liposomes, respectively. Our investigations demonstrated that fusion is a multifactorial process influenced by the chemical composition of the liposomes, the liposomal surface charge, and components of the microbial cell envelope. Penetration and retention within preformed S. aureus biofilms were assessed for liposomes with various surface charges. All liposomes, regardless of surface charge, were capable of penetrating and diffusing through the biofilm matrix. However, cationic liposomes displayed greatest interaction and retention. Biocompatibility was confirmed through haemolysis and cytotoxicity studies. The cholesterol-free fusogenic liposomes developed in this study demonstrated promising potential as drug delivery systems for incorporating antimicrobial agents for biofilm treatment.
Collapse
Affiliation(s)
- Ahmed M Amer
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway.
| | - Colin Charnock
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway
| | - Sanko Nguyen
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway
| |
Collapse
|
24
|
Azra, Khan TA, Ul Haq I, Hinthong W, Campino S, Gohar A, Khan N, Kashif M, Ullah I, Clark TG. Antibiotic Susceptibility Patterns and Virulence Profiles of Classical and Hypervirulent Klebsiella pneumoniae Strains Isolated from Clinical Samples in Khyber Pakhtunkhwa, Pakistan. Pathogens 2025; 14:79. [PMID: 39861040 PMCID: PMC11768992 DOI: 10.3390/pathogens14010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The emergence of hypervirulent and carbapenem-resistant hypermucoviscous Klebsiella pneumoniae strains presents a significant public health challenge due to their increased virulence and resistance to multiple antibiotics. This study evaluates the antibiotic susceptibility patterns and virulence profiles of classical and hypervirulent K. pneumoniae strains isolated from various clinical samples. A total of 500 clinical samples were collected from patients at the Mardan Medical Complex and Ayub Medical Complex in KPK between July 2022 and June 2024. Among these, 64 K. pneumoniae strains were isolated and subsequently subjected to antimicrobial susceptibility testing (AST) and phenotypic virulence detection. Among the 64 isolates, 21 (32.8%) exhibited hypermucoviscosity, a characteristic associated with increased pathogenicity. Hemagglutination was observed in 35 (54.1%) of the isolates, indicating the presence of surface adhesins that facilitate bacterial adherence to host tissues. A high prevalence of biofilm formation was noted, with 54 (84%) isolates capable of forming biofilms, which are known to protect bacteria from antibiotics and the host immune response. Most isolates (59/64, 92.1%) were resistant against ampicillin, highlighting its limited efficacy against these strains. Conversely, the lowest resistance was observed for tigecycline, with only 15% (10/64) of the isolates showing resistance, indicating its potential utility as a treatment option. The study also found that 38 (59.3%) of the isolates were extended-spectrum beta-lactamase (ESBL) producers, 42 (65.6%) were multidrug-resistant (MDR), 32 (50%) were extensively drug-resistant (XDR), and 13 (20.3%) were resistant to carbapenems. The genetic study revealed biofilm producer and enhancer genes (mrkD, pgaABCD, fimH, treC, wzc, pilQ, and luxS) mainly in the hypervirulent strains. These hypervirulent strains also show a high number of resistance genes. The findings of this study underscore the critical need for the active surveillance of antimicrobial resistance and virulence determinants in K. pneumoniae. The coexistence of high levels of antibiotic resistance and virulence factors in these isolates poses a severe threat to public health, as it can lead to difficult-to-treat infections and increased morbidity and mortality.
Collapse
Affiliation(s)
- Azra
- Institute of Pathology and Diagnostic Medicines, IPDM, Khyber Medical University Peshawar, Peshawar 25000, Pakistan; (A.); (T.A.K.)
| | - Taj Ali Khan
- Institute of Pathology and Diagnostic Medicines, IPDM, Khyber Medical University Peshawar, Peshawar 25000, Pakistan; (A.); (T.A.K.)
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland;
- Joint Doctoral School, Silesian University of Technology, 44-100 Gliwice, Poland
- Postgraduate Program in Technological Innovation, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Woranich Hinthong
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (W.H.); (S.C.)
| | - Susana Campino
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (W.H.); (S.C.)
- Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Aisha Gohar
- Microbiology Department, Bacha Khan Medical College Mardan, Mardan 23200, Pakistan;
| | - Noman Khan
- Al Rasheed Hospital & Kidney Center, Opposite Gilani Mart, Maneshra Road, Abbottabad 22020, Pakistan;
| | - Muhammad Kashif
- Public Health Reference Laboratory, Khyber Medical University Peshawar, Peshawar 25000, Pakistan
| | - Ihsan Ullah
- Institute of Pathology and Diagnostic Medicines, IPDM, Khyber Medical University Peshawar, Peshawar 25000, Pakistan; (A.); (T.A.K.)
| | - Taane G. Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (W.H.); (S.C.)
| |
Collapse
|
25
|
Gołasz P, Płoska A, Korniienko V, Diedkova K, Varava Y, Zieliński R, Pogorielov M, Simka W. Modification of Ti13Nb13Zr Alloy Surface via Plasma Electrolytic Oxidation and Silver Nanoparticles Decorating. MATERIALS (BASEL, SWITZERLAND) 2025; 18:349. [PMID: 39859817 PMCID: PMC11767042 DOI: 10.3390/ma18020349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
The dynamically developing field of implantology requires researchers to search for new materials and solutions. In this study, TiNbZr samples were investigated as an alternative for popular, but potentially hazardous TiAl6V4. Samples were etched, sandblasted, subjected to PEO, and covered in AgNP suspension. Simultaneously, SEM images were taken, and the wettability and roughness of the surface were measured. Samples covered in AgNPs were subjected to biological trials. A six-day measurement of human fibroblast proliferation was conducted to assess biocompatibility, and the population of E. coli and S. aureus was measured over eight hours. Results showed that the TiNbZr PEO surface is biocompatible with human fibroblast cells and promotes growth. However, deposited AgNPs exhibited only slight effectiveness in decreasing bacterial growth over the first two hours. The results suggest that the method of surface preparation is sufficient and might promote osseointegration. On the other hand, more efficient and reliable methods of application of AgNPs should be researched.
Collapse
Affiliation(s)
- Przemysław Gołasz
- Department of Inorganic Chemistry, Analytical Chemistry, and Electrochemistry, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland; (P.G.); (A.P.); (M.P.)
- Chemistry Students Research Society, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Agnieszka Płoska
- Department of Inorganic Chemistry, Analytical Chemistry, and Electrochemistry, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland; (P.G.); (A.P.); (M.P.)
| | - Viktoriia Korniienko
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (V.K.); (K.D.)
- Biomedical Research Centre, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
| | - Kateryna Diedkova
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (V.K.); (K.D.)
- Biomedical Research Centre, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
| | - Yuliia Varava
- Biomedical Research Centre, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
| | | | - Maksym Pogorielov
- Department of Inorganic Chemistry, Analytical Chemistry, and Electrochemistry, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland; (P.G.); (A.P.); (M.P.)
| | - Wojciech Simka
- Department of Inorganic Chemistry, Analytical Chemistry, and Electrochemistry, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland; (P.G.); (A.P.); (M.P.)
| |
Collapse
|
26
|
Dafinone ME, Lyle RE, Lee C, Mehta A, Dahle SE, Isseroff RR. Non-antibiotic approaches to mitigating wound infections: Potential for SSRIs and adrenergic antagonists as emerging therapeutics. Wound Repair Regen 2025; 33:e13240. [PMID: 39737521 DOI: 10.1111/wrr.13240] [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: 04/05/2024] [Revised: 10/16/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025]
Abstract
Bacterial biofilms represent a formidable challenge in the treatment of chronic wounds, largely because of their resistance to conventional antibiotics. The emergence of multidrug-resistant (MDR) bacterial strains exacerbates this issue, necessitating a shift towards exploring alternative therapeutic approaches. In response to this urgent need, there has been a surge in research efforts aimed at identifying effective non-antibiotic treatments. Recently noted among the non-antibiotic options are selective serotonin reuptake inhibitors (SSRIs) and beta-adrenergic (β-AR) antagonists. Both have demonstrated antimicrobial activities and wound-healing properties, which makes them particularly promising potential therapeutics for chronic wounds. This review seeks to comprehensively evaluate the landscape of non-antibiotic strategies for managing wound infections. By analysing the latest research findings and clinical developments, it aims to shed light on emerging therapeutic alternatives. Additionally, the review delves into the potential of repurposing systemic therapeutics for topical application, offering insights into the feasibility and challenges associated with current approaches. We also address the necessity of translating promising preclinical results into tangible clinical benefits.
Collapse
Affiliation(s)
- Mirabel E Dafinone
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- School of Medicine, University of Nevada Reno, Reno, Nevada, USA
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
| | - Rawlings E Lyle
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
- School of Medicine, University of California Davis, Davis, California, USA
| | - Conan Lee
- School of Medicine, University of California Davis, Davis, California, USA
| | - Alisha Mehta
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
- College of Medicine, California Northstate University, Elk Grove, California, USA
| | - Sara E Dahle
- School of Medicine, University of California Davis, Davis, California, USA
- Podiatry Section, VA Northern California Health Care System, McClellan Park, California, USA
| | - R Rivkah Isseroff
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
| |
Collapse
|
27
|
Patra S, Saha S, Singh R, Tomar N, Gulati P. Biofilm battleground: Unveiling the hidden challenges, current approaches and future perspectives in combating biofilm associated bacterial infections. Microb Pathog 2025; 198:107155. [PMID: 39586337 DOI: 10.1016/j.micpath.2024.107155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
A biofilm is a complex aggregation of microorganisms, either of the same or different species, that adhere to a surface and are encased in an extracellular polymeric substances (EPS) matrix. Quorum sensing (QS) and biofilm formation are closely linked, as QS genes regulate the development, maturation, and breakdown of biofilms. Inhibiting QS can be utilized as an effective approach to combat the impacts of biofilm infection. The impact of biofilms includes chronic infections, industrial biofouling, infrastructure corrosion, and environmental contamination as well. Therefore, a deep understanding of biofilms is crucial for enhancing public health, advancing industrial processes, safeguarding the environment, and deepening our knowledge of microbial life as well. This review aims to offer a comprehensive examination of challenges posed by bacterial biofilms, contemporary approaches and strategies for effectively eliminating biofilms, including the inhibition of quorum sensing pathways, while also focusing on emerging technologies and techniques for biofilm treatment. In addition, future research is projected to target the challenges associated with the bacterial biofilms, striving to develop new approaches and improve existing strategies for their effective control and eradication.
Collapse
Affiliation(s)
- Sandeep Patra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sumana Saha
- Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Randhir Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Nandini Tomar
- Department of Biotechnology, South Asian University, New Delhi, India
| | - Pallavi Gulati
- Ram Lal Anand College, University of Delhi, New Delhi, India.
| |
Collapse
|
28
|
Timmis K, Karahan ZC, Ramos JL, Koren O, Pérez‐Cobas AE, Steward K, de Lorenzo V, Caselli E, Douglas M, Schwab C, Rivero V, Giraldo R, Garmendia J, Turner RJ, Perlmutter J, Borrero de Acuña JM, Nikel PI, Bonnet J, Sessitsch A, Timmis JK, Pruzzo C, Prieto MA, Isazadeh S, Huang WE, Clarke G, Ercolini D, Häggblom M. Microbes Saving Lives and Reducing Suffering. Microb Biotechnol 2025; 18:e70068. [PMID: 39844583 PMCID: PMC11754571 DOI: 10.1111/1751-7915.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology and Ibn‐i Sina Hospital Central Microbiology LaboratoryAnkara University School of MedicineAnkaraTurkey
| | - Juan Luis Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del ZaidínGranadaSpain
| | - Omry Koren
- Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Ana Elena Pérez‐Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS)Ramón y Cajal University HospitalMadridSpain
- CIBER in Infectious Diseases (CIBERINFEC)MadridSpain
| | | | - Victor de Lorenzo
- Department of Systems BiologyNational Centre of Biotechnology CSICMadridSpain
| | - Elisabetta Caselli
- Section of Microbiology, Department of Environmental and Prevention SciencesUniversity of FerraraFerraraItaly
| | - Margaret Douglas
- Usher InstituteUniversity of Edinburgh Medical School, and Public Health ScotlandEdinburghUK
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Virginia Rivero
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Rafael Giraldo
- Department of Microbial BiotechnologyNational Centre for Biotechnology (CNB‐CSIC)MadridSpain
| | - Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de Navarra, MutilvaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Raymond J. Turner
- Department of Biological SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | | | - Pablo Ivan Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkLyngbyDenmark
| | - Jerome Bonnet
- Centre de Biochimie Structurale, INSERM/CNRSUniversity of MontpellierMontpellierFrance
| | - Angela Sessitsch
- Bioresources UnitAIT Austrian Institute of TechnologyViennaAustria
| | - James K. Timmis
- Department of Political ScienceUniversity of FreiburgFreiburgGermany
- Athena Institute for Research on Innovation and Communication in Health and Life SciencesVrije UniversiteitAmsterdamThe Netherlands
| | - Carla Pruzzo
- Department of Earth, Environmental and Life Sciences (DISTAV)University of GenoaGenovaItaly
| | - M. Auxiliadora Prieto
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Siavash Isazadeh
- Corporate Technical & PerformanceVeolia North AmericaParamusNew JerseyUSA
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry & Neurobehavioral SciencesUniversity College CorkCorkIreland
| | - Danilo Ercolini
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Max Häggblom
- Department of Biochemistry and Microbiology, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
29
|
Syed AK, Baral R, Van Vlack ER, Gil-Marqués ML, Lenhart T, Hooper DC, Kahne D, Losick R, Bradshaw N. Biofilm formation by Staphylococcus aureus is triggered by a drop in the levels of a cyclic dinucleotide. Proc Natl Acad Sci U S A 2024; 121:e2417323121. [PMID: 39680756 PMCID: PMC11670122 DOI: 10.1073/pnas.2417323121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
The bacterial pathogen Staphylococcus aureus forms multicellular communities known as biofilms in which cells are held together by an extracellular matrix principally composed of repurposed cytoplasmic proteins and extracellular DNA. These biofilms assemble during infections or under laboratory conditions by growth on medium containing glucose, but the intracellular signal for biofilm formation and its downstream targets were unknown. Here, we present evidence that biofilm formation is triggered by a drop in the levels of the second messenger cyclic-di-AMP. Previous work identified genes needed for the release of extracellular DNA, including genes for the cyclic-di-AMP phosphodiesterase GdpP, the transcriptional regulator XdrA, and the purine salvage enzyme Apt. Using a cyclic-di-AMP riboswitch biosensor and mass spectrometry, we show that the second messenger drops in abundance during biofilm formation in a glucose-dependent manner. Mutation of these three genes elevates cyclic-di-AMP and prevents biofilm formation in a murine catheter model. Supporting the generality of this mechanism, we found that gdpP was required for biofilm formation by diverse strains of S. aureus. We additionally show that the downstream consequence of the drop in cyclic-di-AMP is inhibition of the "accessory gene regulator" operon agr, which is known to suppress biofilm formation through phosphorylation of the transcriptional regulator AgrA by the histidine kinase AgrC. Consistent with this, an agr mutation bypasses the block in biofilm formation and eDNA release caused by a gdpP mutation. Finally, we report the unexpected observation that GdpP inhibits phosphotransfer from AgrC to AgrA, revealing a direct connection between the phosphodiesterase and agr.
Collapse
Affiliation(s)
- Adnan K. Syed
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA02138
| | - Rishika Baral
- Department of Biochemistry, Brandeis University, Waltham, MA02453
| | - Erik R. Van Vlack
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA02138
| | | | - Taliesin Lenhart
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA02138
| | - David C. Hooper
- Division of Infectious Disease, Massachusetts General Hospital, Boston, MA02114
| | - Daniel Kahne
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA02138
| | - Richard Losick
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA02138
| | - Niels Bradshaw
- Department of Biochemistry, Brandeis University, Waltham, MA02453
| |
Collapse
|
30
|
Zahorska E, Denig LM, Lienenklaus S, Kuhaudomlarp S, Tschernig T, Lipp P, Munder A, Gillon E, Minervini S, Verkhova V, Imberty A, Wagner S, Titz A. High-Affinity Lectin Ligands Enable the Detection of Pathogenic Pseudomonas aeruginosa Biofilms: Implications for Diagnostics and Therapy. JACS AU 2024; 4:4715-4728. [PMID: 39735928 PMCID: PMC11672137 DOI: 10.1021/jacsau.4c00670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/31/2024]
Abstract
Pseudomonas aeruginosa is a critical priority pathogen and causes life-threatening acute and biofilm-associated chronic infections. The choice of suitable treatment for complicated infections requires lengthy culturing for species identification from swabs or an invasive biopsy. To date, no fast, pathogen-specific diagnostic tools for P. aeruginosa infections are available. Here, we present the noninvasive pathogen-specific detection of P. aeruginosa using novel fluorescent probes that target the bacterial biofilm-associated lectins LecA and LecB. Several glycomimetic probes were developed to target these extracellular lectins and demonstrated to stain P. aeruginosa biofilms in vitro. Importantly, for the targeting of LecA an activity boost to low-nanomolar affinity could be achieved, which is essential for in vivo application. In vitro, the nanomolar divalent LecA-targeted imaging probe accumulated effectively in biofilms under flow conditions, independent of the fluorophore identity. Investigation of these glycomimetic imaging probes in a murine lung infection model and fluorescence imaging revealed accumulation at the infection site. These findings demonstrate the use of LecA- and LecB-targeting probes for the imaging of P. aeruginosa infections and suggest their potential as pathogen-specific diagnostics to accelerate the start of the appropriate treatment.
Collapse
Affiliation(s)
- Eva Zahorska
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Lisa Marie Denig
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Stefan Lienenklaus
- Hannover
Medical School, Institute of Laboratory
Animal Science, Hannover 30625, Germany
| | - Sakonwan Kuhaudomlarp
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
- Department
of Biochemistry, Faculty of Science, Mahidol
University, Bangkok 10400, Thailand
- Center
for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Thomas Tschernig
- Medical
Faculty of Saarland University, Institute
of Anatomy and Cell Biology, Homburg/Saar, D-66421, Germany
| | - Peter Lipp
- Center
for Molecular Signaling (PZMS), Medical
Faculty of Saarland University, Homburg/Saar D-66421, Germany
| | - Antje Munder
- Department
of Pediatric Pneumology, Allergology and
Neonatology, Hannover Medical School, Carl Neuberg-Str. 1, Hannover D-30625, Germany
- Biomedical
Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover D-30625, Germany
| | - Emilie Gillon
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
| | - Saverio Minervini
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
| | - Varvara Verkhova
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Anne Imberty
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
| | - Stefanie Wagner
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Alexander Titz
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| |
Collapse
|
31
|
Karrar Alsharif MH, Poyil MM, Bin Dayel S, Alqahtani MS, Albadrani AA, Omar ZMM, Arafah AMR, Alarabi TGM, Fayyad RM, Abd El-Lateef AELS. Eradication of Biofilms on Catheters: Potentials of Tamarix ericoides Rottl. Bark Coating in Preventing Catheter-Associated Urinary Tract Infections (CAUTIs). Life (Basel) 2024; 14:1593. [PMID: 39768301 PMCID: PMC11677003 DOI: 10.3390/life14121593] [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: 11/04/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Catheter-associated urinary tract infections (CAUTIs) cause serious complications among hospitalized patients due to biofilm-forming microorganisms which make treatment ineffective by forming antibiotic-resistant strains. As most CAUTI-causing bacterial pathogens have already developed multidrug resistance, there is an urgent need for alternative antibacterial agents to prevent biofilms on catheter surfaces. As a trial to find out such a potential agent of natural origin, the bark of Tamarix ericoides Rottl., a little-known plant from the Tamaricaceae family, was examined for its antibacterial and antibiofilm activities against one of the major, virulent, CAUTI-causing bacterial pathogens: Enterococcus faecalis. The methanolic T. ericoides bark extract was analyzed for its antibacterial activity using the well diffusion method and microdilution method. Killing kinetics were calculated using time-kill assay, and the ability of biofilm formation and its eradication upon treatment with the T. ericoides bark extract was studied by crystal violet assay. GC-MS analysis was performed to understand the phytochemical presence in the extract. A in vitro bladder model study was performed using extract-coated catheters against E. faecalis, and the effect was visualized using CLSM. The changes in the cell morphology of the bacterium after treatment with the T. ericoides bark extract were observed using SEM. The biocompatibility of the extract towards L929 cells was studied by MTT assay. The anti-E. faecalis activity of the extract-coated catheter tube was quantified by viable cell count method, which exposed 20% of growth after five days of contact with E. faecalis. The anti-adhesive property of the T. ericoides bark extract was studied using CLSM. The extract showed potential antibacterial activity, and the lowest inhibitory concentration needed to inhibit the growth of E. faecalis was found to be 2 mg/mL. The GC-MS analysis of the methanolic fractions of the T. ericoides bark extract revealed the presence of major phytochemicals, such as diethyl phthalate, pentadecanoic acid, methyl 6,11-octadecadienoate, cyclopropaneoctanoic acid, 2-[(2-pentylcyclopropyl) methyl]-, methyl ester, erythro-7,8-bromochlorodisparlure, etc., that could be responsible for the antibacterial activity against E. faecalis. The killing kinetics of the extract against E. faecalis was calculated and the extract showed promising antibiofilm activity on polystyrene surfaces. The T. ericoides bark extract effectively reduced the E. faecalis mature biofilms by 75%, 82%, and 83% after treatment with 1X MIC (2 mg/mL), 2X MIC (4 mg/mL), and 3X MIC (6 mg/mL) concentrations, respectively, which was further confirmed by SEM analysis. The anti-adhesive property of the T. ericoides bark extract studied using CLSM revealed a reduction in the biofilm thickness, and the FDA and PI combination revealed the death of 80% of the cells on the extract-coated catheter tube. In addition, SEM analysis showed extensive damage to the E. faecalis cells after the T. ericoides bark extract treatment, and it was not cytotoxic. Hence, after further studies, T. ericoides bark extract with potential antibacterial, antibiofilm, and anti-adhesive activities can be developed as an alternative agent for treating CAUTIs.
Collapse
Affiliation(s)
- Mohammed H. Karrar Alsharif
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.M.P.); (Z.M.M.O.); (A.M.A.); (A.E.-L.S.A.E.-L.)
| | - Muhammad Musthafa Poyil
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.M.P.); (Z.M.M.O.); (A.M.A.); (A.E.-L.S.A.E.-L.)
| | - Salman Bin Dayel
- Department of Internal Medicine, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (S.B.D.); (M.S.A.); (A.A.A.)
| | - Mohammed Saad Alqahtani
- Department of Internal Medicine, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (S.B.D.); (M.S.A.); (A.A.A.)
| | - Ahmed Abdullah Albadrani
- Department of Internal Medicine, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (S.B.D.); (M.S.A.); (A.A.A.)
| | - Zainab Mohammed M. Omar
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.M.P.); (Z.M.M.O.); (A.M.A.); (A.E.-L.S.A.E.-L.)
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Assiut 71524, Egypt
| | - Abdullah MR. Arafah
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.M.P.); (Z.M.M.O.); (A.M.A.); (A.E.-L.S.A.E.-L.)
| | | | - Reda M. Fayyad
- Department Pharmacology, General Medicine Practice Program, Batterjee Medical College, Asser 61961, Saudi Arabia;
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo 11511, Egypt
| | - Abd El-Lateef Saeed Abd El-Lateef
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.M.P.); (Z.M.M.O.); (A.M.A.); (A.E.-L.S.A.E.-L.)
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo 11511, Egypt
| |
Collapse
|
32
|
Sharma A, Kumar Y, Kumar G, Tahlan AK. Biofilm Production and Antibiogram Profiles in Escherichia coli and Salmonella. Indian J Microbiol 2024; 64:1512-1517. [PMID: 39678983 PMCID: PMC11645373 DOI: 10.1007/s12088-023-01149-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2024] Open
Abstract
Salmonella and Escherichia coli are important enteric pathogens associated with a variety of infections. Biofilm formation and antimicrobial resistance are important characteristics making these pathogens a concern in terms of strong attachment to substrates, expression of virulence markers and difficult removal. The present study investigates the biofilm-forming ability and antibiogram patterns among E. coli and Salmonella spp. A total of 200 E. coli and 100 Salmonella isolates received at National Salmonella and Escherichia Centre were identified by biochemical testing, followed by serotyping. Biofilm production was detected by Tissue Culture Plate method. The isolates were further subjected to Antibiotic Susceptibility Testing by the Kirby-Bauer disk diffusion method. 113 (56.5%) E. coli isolates and 79 (79%) Salmonella isolates were detected as biofilm producers. A total of 114(57%)E. coli isolates and 31(31%) Salmonella isolates were found to be resistant to multiple drugs when Antibiotic Susceptibility Testing was carried out. Antibiotic resistance was found to be significantly higher in biofilm producing salmonella (p = 0.001) whereas in the case of E. coli the difference remained non-significant (p = 0.4454). The capability to produce biofilm along with acquiring high level of antimicrobial resistance in salmonella and E. coli provide enhanced survival potential in adverse environments. Therefore, it becomes a serious cause of concern for public health authorities considering the virulence of these bacteria and their association with different disease conditions and requires urgent intervention with regards to control and prevention strategies.
Collapse
Affiliation(s)
- A. Sharma
- National Salmonella and Escherichia Centre, Central Research Institute, Kasauli, Himachal Pradesh 173204 India
| | - Y. Kumar
- National Salmonella and Escherichia Centre, Central Research Institute, Kasauli, Himachal Pradesh 173204 India
| | - G. Kumar
- National Salmonella and Escherichia Centre, Central Research Institute, Kasauli, Himachal Pradesh 173204 India
| | - A. K. Tahlan
- National Salmonella and Escherichia Centre, Central Research Institute, Kasauli, Himachal Pradesh 173204 India
| |
Collapse
|
33
|
Yoshida S, Inaba H, Nomura R, Nakano K, Matsumoto-Nakano M. Role of fimbriae variations in Porphyromonas gulae biofilm formation. J Oral Biosci 2024; 66:28-33. [PMID: 39216533 DOI: 10.1016/j.job.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Porphyromonas gulae is a major causative agent of periodontal disease in companion animals that possesses various virulence factors, including fimbriae, lipopolysaccharides, and proteases. P. gulae fimbriae are classified into three genotypes (A, B, and C) based on their nucleotide sequences. Type C fimbrial isolates have been reported to be more virulent than other fimA types, suggesting that different fimA types may aid in the regulation of periodontal pathogenesis. Detailed findings regarding the ability of P. gulae to form biofilms have yet to be reported. Here, we investigated the contributions of fimbrial genotypes in P. gulae biofilm formation. METHODS P. gulae and P. gingivalis biofilms were generated on plates and analyzed using confocal laser microscopy. Additionally, the biofilms formed were assessed by staining with crystal violet. Furthermore, the physical strength of P. gulae biofilms was examined by ultrasonication. RESULTS Biofilms formed by P. gulae type C were denser than those formed by types A and B. Moreover, the amount of biofilm formed by type C strains was significantly greater than that formed by type A and B strains, which was similar to the biofilms formed by P. gingivalis with type II fimbriae. Additionally, the physical strength of the type C biofilm was significantly greater than that of the other strains. CONCLUSIONS These results suggest that FimA variation may coordinate for biofilm formation. This is the first report on the observation and characterization of P. gulae biofilm formation.
Collapse
Affiliation(s)
- Sho Yoshida
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Inaba
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Department of Dental Hygiene, Kyoto Koka Women's College, Kyoto, Japan.
| | - Ryota Nomura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Michiyo Matsumoto-Nakano
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
34
|
Xi Y, Li X, Liu L, Xiu F, Yi X, Chen H, You X. Sneaky tactics: Ingenious immune evasion mechanisms of Bartonella. Virulence 2024; 15:2322961. [PMID: 38443331 PMCID: PMC10936683 DOI: 10.1080/21505594.2024.2322961] [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: 12/18/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Gram-negative Bartonella species are facultative intracellular bacteria that can survive in the harsh intracellular milieu of host cells. They have evolved strategies to evade detection and degradation by the host immune system, which ensures their proliferation in the host. Following infection, Bartonella alters the initial immunogenic surface-exposed proteins to evade immune recognition via antigen or phase variation. The diverse lipopolysaccharide structures of certain Bartonella species allow them to escape recognition by the host pattern recognition receptors. Additionally, the survival of mature erythrocytes and their resistance to lysosomal fusion further complicate the immune clearance of this species. Certain Bartonella species also evade immune attacks by producing biofilms and anti-inflammatory cytokines and decreasing endothelial cell apoptosis. Overall, these factors create a challenging landscape for the host immune system to rapidly and effectively eradicate the Bartonella species, thereby facilitating the persistence of Bartonella infections and creating a substantial obstacle for therapeutic interventions. This review focuses on the effects of three human-specific Bartonella species, particularly their mechanisms of host invasion and immune escape, to gain new perspectives in the development of effective diagnostic tools, prophylactic measures, and treatment options for Bartonella infections.
Collapse
Affiliation(s)
- Yixuan Xi
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Xinru Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Lu Liu
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Feichen Xiu
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Xinchao Yi
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Hongliang Chen
- Chenzhou NO.1 People’s Hospital, The Affiliated Chenzhou Hospital, Hengyang Medical College, University of South China, ChenZhou, China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
35
|
Wu Z, Cai Y, Han Y, Su Y, Zhang T, Wang X, Yan A, Wang L, Wu S, Wang G, Zhang Z. Development of α-Helical Antimicrobial Peptides with Imperfect Amphipathicity for Superior Activity and Selectivity. J Med Chem 2024; 67:19561-19572. [PMID: 39484706 DOI: 10.1021/acs.jmedchem.4c01855] [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: 11/03/2024]
Abstract
The advancement of antimicrobial peptides (AMPs) as therapeutic agents is hindered by their poor selectivity. Recent evidence indicates that controlled disruption of the amphipathicity of α-helical AMPs may increase the selectivity. This study investigated the role of imperfect amphipathicity in optimizing AMPs with varied sequences to enhance their activity and selectivity. Among these, the lead peptide RI-18, characterized by an imperfectly amphipathic α-helical structure, demonstrated potent and broad-spectrum antibacterial activity without inducing hemolytic or cytotoxic effects. RI-18 effectively eliminated planktonic and biofilm-associated bacteria as well as persister cells and exhibited high bacterial plasma membrane affinity, inducing rapid membrane permeabilization and rupture. Notably, RI-18 significantly reduced bacterial loads without promoting bacterial resistance, highlighting its therapeutic potential. Overall, this study identified RI-18 as a promising antimicrobial candidate. The rational strategy of tuning imperfect amphipathicity to enhance the AMP activity and selectivity may facilitate the design and development of AMPs.
Collapse
Affiliation(s)
- Zhongxiang Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
| | - Ying Cai
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
| | - Yajun Han
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yunhan Su
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Tianyu Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
| | - Xingyu Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - An Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
| | - Liunan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
| | - Sijing Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Gan Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, China
| | - Zhiye Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650031, China
| |
Collapse
|
36
|
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: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [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
|
37
|
Kulayta K, Zerdo Z, Seid M, Dubale A, Manilal A, Kebede T, Alahmadi RM, Raman G, Akbar I. Biofilm formation and antibiogram profile of bacteria from infected wounds in a general hospital in southern Ethiopia. Sci Rep 2024; 14:26359. [PMID: 39487302 PMCID: PMC11530625 DOI: 10.1038/s41598-024-78283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
Biofilm-producing bacteria associated with wound infections exhibit exceptional drug resistance, leading to an escalation in morbidity, worse clinical outcomes (including delay in the healing process), and an increase in health care cost, burdening the whole system. This study is an attempt to estimate the prevalence and the relationship between the biofilm-forming capacity and multi-drug resistance of wound bacterial isolates. The findings intended to help clinicians, healthcare providers and program planners and to formulate an evidence-based decision-making process, especially in resource-limited healthcare settings. This study was done to assess the prevalence of bacterial infections in wounds and the antibiogram and biofilm-forming capacity of those bacteria in patients with clinical signs and symptoms, attending a General Hospital in southern Ethiopia. A cross-sectional study was performed in Arba Minch General Hospital from June to November 2021. The study participants comprised 201 patients with clinically infected wounds. Demographic and clinical data were gathered via a structured questionnaire. Specimens from wounds were taken from each participant and inoculated onto a series of culture media, namely MacConkey agar, mannitol salt agar, and blood agar, and different species were identified using a number of biochemical tests. Antimicrobial susceptibility tests were performed by means of the Kirby-Bauer disc diffusion technique following the guidelines of the Clinical and Laboratory Standards Institute. A micro-titer plate method was employed to detect the extent of biofilm formation. Bivariable and multivariable logistic regression models were applied to analyse the association between dependent and independent variables, and P values ≤ 0.05 were considered as statistically significant. Data analyses were done with Statistical Package for the Social Sciences version 25. Out of the 201 clinically infected wounds, 165 were found culture-positive with an overall prevalence of 82% (95% CI: 75.9-86.9). In total, 188 bacteria were recovered; 53.1% of them were Gram-positive cocci. The often-isolated bacterial species were Staphylococcus aureus, 38.3% (n = 72), and Pseudomonas aeruginosa, 16.4% (n = 31). The Gram-positive isolates showed considerable resistance against penicillin, 70%, and somewhat strong resistance against tetracycline, 57.7%. Gram-negative isolates showed severe resistance to ampicillin, 80.68%. The overall multi-drug resistance (MDR) among isolates was 48.4%. Extended beta-lactamase (ESBL)-producing Gram-negatives and methicillin-resistant Staphylococcus aureus (MRSA) accounted for 49 and 41.67%, respectively; 62.2% of the isolates were biofilm formers and were correlated statistically with MDR, ESBL producers, and MRSA (P < 0.005). The extent of biofilm formation and the prevalence of MDR bacteria associated with infected wounds hint at a public health threat that needs immediate attention. Thus, a more balanced and comprehensive wound management approach and antimicrobial stewardship program are essential in the study setting.
Collapse
Affiliation(s)
- Kebede Kulayta
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
- Department of Medical Laboratory Science, Arba Minch College of Health Sciences, Arba Minch, Ethiopia
| | - Zerihun Zerdo
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
| | - Mohammed Seid
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia.
| | - Amanuel Dubale
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
| | - Aseer Manilal
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia.
| | - Teshome Kebede
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
- Arba Minch General Hospital, Arba Minch, Ethiopia
| | - Reham M Alahmadi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box 2455, 11451, Riyadh, Saudi Arabia
| | - Gurusamy Raman
- Department of Life Sciences, Yeungnam University, Gyeongsan, Gyeongbuk-Do, South Korea.
| | - Idhayadhulla Akbar
- Research Department of Chemistry, Nehru Memorial College (Affiliated to Bharathidasan University), Puthanampatti, Tiruchirappalli District, 621007, Tamil Nadu, India
| |
Collapse
|
38
|
Chegini Z, Shariati A, Alikhani MY, Safaiee M, Rajaeih S, Arabestani M, Azizi M. Antibacterial and antibiofilm activity of silver nanoparticles stabilized with C-phycocyanin against drug-resistant Pseudomonas aeruginosa and Staphylococcus aureus. Front Bioeng Biotechnol 2024; 12:1455385. [PMID: 39524122 PMCID: PMC11544008 DOI: 10.3389/fbioe.2024.1455385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Biofilms are bacterial communities that can protect them against external factors, including antibiotics. In this study, silver nanoparticles (AgNPs) were formed by modifying AgNPs with C-phycocyanin (Ag-Pc) to inhibit the growth of carbapenem-resistant Pseudomonas aeruginosa (CR P. aeruginosa) and methicillin-resistant Staphylococcus aureus (MRSA) and destroy biofilm of these bacteria. Methods The AgNPs were prepared with the green synthesis method, and Pc was used to stabilize the AgNPs. The Ag-Pc's antibacterial and antibiofilm effects were evaluated using the Microbroth dilution method and microtiter plate assay. The inhibitory effect of Ag-Pc on the expression of biofilm-related genes was evaluated by real-time PCR. Moreover, the MTT assay was used to assess the Ag-Pc toxicity. Results The Ag-Pc minimum inhibitory concentration (MIC) was 7.4 μg/mL for CR P. aeruginosa and MRSA. Pc did not show antibacterial effects against any of the strains. Ag-Pc suppressed biofilm formation and destroyed matured biofilm in both bacteria more efficiently than the AgNPs (P< 0.05). The expression of all genes was not significantly reduced in the presence of synthesized nanoparticles. Finally, the MTT assay results did not show toxicity against a murine fibroblast cell line (L929) at MIC concentration. Conclusion The present study showed the promising potential of Pc for improving the antibacterial and antibiofilm function of AgNPs and inhibiting drug-resistant bacteria. Therefore, Ag-Pc nanoparticles can be considered a promising therapeutic approach for the managing of the bacterial biofilm.
Collapse
Affiliation(s)
- Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of medical sciences, Arak, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maliheh Safaiee
- Department of Organic Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Shahin Rajaeih
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Infectious Diseases Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Regenerative Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
39
|
Perini HF, Pereira BDB, Sousa EG, Matos BS, Silva Prado LCD, Carvalho Azevedo VAD, Castro Soares SD, Silva MVD. Inhibitory effect of Bacillus velezensis 1273 strain cell-free supernatant against developing and preformed biofilms of Staphylococcus aureus and MRSA. Microb Pathog 2024; 197:107065. [PMID: 39447663 DOI: 10.1016/j.micpath.2024.107065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Microbial biofilms constitute a significant virulence factor and a substantial challenge in clinical environments due to their role in promoting antimicrobial resistance and their resilience to eradication efforts. Methicillin-resistant Staphylococcus aureus (MRSA) infections substantially increase healthcare costs, extend hospitalizations, and elevate morbidity and mortality rates. Therefore, developing innovative strategies to target and eliminate these bacteria and their biofilms effectively is imperative for robust epidemiological control. In this study, we evaluated the antibacterial and antibiofilm activities of cell-free supernatant (CFS) obtained from the Bacillus velezensis 1273 strain culture. Our data showed that CFS inhibited the growth of S. aureus ATCC 29213 and MRSA (clinical strain), with greater efficacy observed against S. aureus (1:16 dilution). Furthermore, CFS showed substantial potential in reducing biofilm formation in both strains (∼30 %) at subinhibitory concentrations. Additionally, the antibacterial activity against biofilm-formed cells showed that pure CFS treatment decreased the viability of S. aureus (60 %) and MRSA (45 %) sessile cells. We further demonstrated that CFS treatment induces the production of reactive oxygen species (ROS) and damages the membranes and cell walls of the pathogen cells. Genome analysis revealed the presence of genes encoding bacteriocins and secondary metabolites with antibacterial activity in the B. velezensis 1273 genome. These findings highlight the potential of probiotic bacterial metabolites as antibiofilm and anti-multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Hugo Felix Perini
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | - Bianca de Barros Pereira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Eduarda Guimarães Sousa
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz Sodré Matos
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | | | - Siomar de Castro Soares
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marcos Vinicius da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
40
|
Liu HY, Prentice EL, Webber MA. Mechanisms of antimicrobial resistance in biofilms. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:27. [PMID: 39364333 PMCID: PMC11445061 DOI: 10.1038/s44259-024-00046-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
Most bacteria in nature exist in aggregated communities known as biofilms, and cells within a biofilm demonstrate major physiological changes compared to their planktonic counterparts. Biofilms are associated with many different types of infections which can have severe impacts on patients. Infections involving a biofilm component are often chronic and highly recalcitrant to antibiotic therapy as a result of intrinsic physical factors including extracellular matrix production, low growth rates, altered antibiotic target production and efficient exchange of resistance genes. This review describes the biofilm lifecycle, phenotypic characteristics of a biofilm, and contribution of matrix and persister cells to biofilms intrinsic tolerance to antimicrobials. We also describe how biofilms can evolve antibiotic resistance and transfer resistance genes within biofilms. Multispecies biofilms and the impacts of various interactions, including cooperation and competition, between species on tolerance to antimicrobials in polymicrobial biofilm communities are also discussed.
Collapse
Affiliation(s)
- Ho Yu Liu
- Quadram Institute Biosciences, Norwich Research Park, Norwich, Norfolk NR4 7UQ UK
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7TJ UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, Norfolk NR4 7UG UK
| | - Emma L Prentice
- Quadram Institute Biosciences, Norwich Research Park, Norwich, Norfolk NR4 7UQ UK
| | - Mark A Webber
- Quadram Institute Biosciences, Norwich Research Park, Norwich, Norfolk NR4 7UQ UK
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7TJ UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, Norfolk NR4 7UG UK
| |
Collapse
|
41
|
Murphy MM, Culligan EP, Murphy CP. Investigating the antimicrobial and antibiofilm properties of marine halophilic Bacillus species against ESKAPE pathogens. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e70027. [PMID: 39446085 PMCID: PMC11500616 DOI: 10.1111/1758-2229.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Antimicrobial resistance (AMR), known as the "silent pandemic," is exacerbated by pathogenic bacteria's ability to form biofilms. Marine compounds hold promise for novel antibacterial drug discovery. Two isolates from preliminary saltwater environment screening demonstrated antimicrobial activity and were subsequently identified as Bacillus subtilis MTUA2 and Bacillus velezensis MTUC2. Minimum inhibitory concentrations (MICs), minimum biofilm inhibition concentrations (MBICs) and minimum biofilm eradication concentrations (MBECs) required to prevent and/or disrupt bacterial growth and biofilm formation were established for MRSA, Staphylococcus aureus, Acinetobacter baumannii and Escherichia coli. The metabolic activity within biofilms was determined by the 2,3,5-triphenyltetrazolium chloride assay. Both Bacillus species exhibited unique antimicrobial effects, reducing MRSA and S. aureus planktonic cell growth by 50% and sessile cell growth for S. aureus and E. coli by 50% and 90%, respectively. No effect was observed against A. baumannii. Significant MBIC and MBEC values were achieved, with 99% inhibition and 90% reduction in MRSA and S. aureus biofilms. Additionally, 90% and 50% inhibition was observed in E. coli and A. baumannii biofilms, respectively, with a 50% reduction in E. coli biofilm. These findings suggest that the mode of action employed by B. subtilis MTUA2 and B. velezensis MTUC2 metabolites should be further characterized and could be beneficial if used independently or in combination with other treatments.
Collapse
Affiliation(s)
- Monica M. Murphy
- Department of Biological SciencesMunster Technological UniversityCorkIreland
| | - Eamonn P. Culligan
- Department of Biological SciencesMunster Technological UniversityCorkIreland
| | - Craig P. Murphy
- Department of Biological SciencesMunster Technological UniversityCorkIreland
| |
Collapse
|
42
|
Shepherd MJ, Fu T, Harrington NE, Kottara A, Cagney K, Chalmers JD, Paterson S, Fothergill JL, Brockhurst MA. Ecological and evolutionary mechanisms driving within-patient emergence of antimicrobial resistance. Nat Rev Microbiol 2024; 22:650-665. [PMID: 38689039 DOI: 10.1038/s41579-024-01041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2024] [Indexed: 05/02/2024]
Abstract
The ecological and evolutionary mechanisms of antimicrobial resistance (AMR) emergence within patients and how these vary across bacterial infections are poorly understood. Increasingly widespread use of pathogen genome sequencing in the clinic enables a deeper understanding of these processes. In this Review, we explore the clinical evidence to support four major mechanisms of within-patient AMR emergence in bacteria: spontaneous resistance mutations; in situ horizontal gene transfer of resistance genes; selection of pre-existing resistance; and immigration of resistant lineages. Within-patient AMR emergence occurs across a wide range of host niches and bacterial species, but the importance of each mechanism varies between bacterial species and infection sites within the body. We identify potential drivers of such differences and discuss how ecological and evolutionary analysis could be embedded within clinical trials of antimicrobials, which are powerful but underused tools for understanding why these mechanisms vary between pathogens, infections and individuals. Ultimately, improving understanding of how host niche, bacterial species and antibiotic mode of action combine to govern the ecological and evolutionary mechanism of AMR emergence in patients will enable more predictive and personalized diagnosis and antimicrobial therapies.
Collapse
Affiliation(s)
- Matthew J Shepherd
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK.
| | - Taoran Fu
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Niamh E Harrington
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Anastasia Kottara
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Kendall Cagney
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Steve Paterson
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Joanne L Fothergill
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Michael A Brockhurst
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
43
|
Rostamani M, Bakht M, Rahimi S, Alizadeh SA, Anari RK, Khakpour M, Javadi A, Fardsanei F, Nikkhahi F. Phenotypic and genotypic determination of resistance to common disinfectants among strains of Acinetobacter baumannii producing and non-producing biofilm isolated from Iran. BMC Microbiol 2024; 24:323. [PMID: 39237859 PMCID: PMC11378455 DOI: 10.1186/s12866-024-03484-1] [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: 05/13/2023] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Nosocomial infections are a global problem in hospitals all around the world. It is considered a major health problem, especially in developing countries. The increase in the patient's stay in hospitals has increased the mortality rate, and consequently, the costs drastically increase. The main purpose of using disinfectants in the hospital environment is to reduce the risk of nosocomial infections. Ethylene diamine tetra acetic acid (EDTA) causes lysis and increases susceptibility to antimicrobial agents in the planktonic form of bacteria. This substance affects the permeability of the outer membrane of bacteria. It also prevents the formation of biofilms by bacteria. MATERIALS AND METHODS In the current study, 120 isolates of Acinetobacter baumannii (A. baumannii) were confirmed by phenotypic and genotypic methods. Antibiogram was performed and then the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of isolates against 5% sodium hypochlorite, ethanol %70, sayasept-HP 2%, chlorhexidine 2%, dettol 4/8% were evaluated. In addition, the disinfectant effect was re-evaluated with the mixture of EDTA solution. All isolates were examined for biofilm presence by crystal violet staining method in triplicates and repeated three times for each strain. Also for all isolates detection of efflux pump genes (Qac-E, qacE-Δ1, SUG-E) by PCR technique was done. RESULTS Antibiogram results of A. baumannii showed that 6.7% were Multi-drug-resistant (MDR), and 89.2% were Extensively drug-resistant (XDR) isolates. The highest effect of disinfectants was related to 5% sodium hypochlorite, and the least effect was 70% ethanol. EDTA increases the efficacy of selected disinfectants significantly. The highest prevalence of the efflux pump genes was related to SUG-E (95%) and Qac-E (91.7%), and, the qacE-Δ1 gene with 12.5%. The biofilm production rate was 91.3% among all isolates. CONCLUSION The best and safest way to disinfect hospital floors and surfaces is to choose the right disinfectants, and learn how to use them properly. In this study, a mixture of disinfectants and EDTA had a significant effect on bactericidal activity. it was found that improper use of disinfectants, especially the use of sub-inhibitory dilutions, increases the resistance of bacteria to disinfectants.
Collapse
Affiliation(s)
- Mohammad Rostamani
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehdi Bakht
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sara Rahimi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Safar Ali Alizadeh
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Raana Kazemzadeh Anari
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohadeseh Khakpour
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amir Javadi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Community Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Fatemeh Fardsanei
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farhad Nikkhahi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
44
|
Liu YS, Zhang C, Khoo BL, Hao P, Chua SL. Dual-species proteomics and targeted intervention of animal-pathogen interactions. J Adv Res 2024:S2090-1232(24)00383-7. [PMID: 39233003 DOI: 10.1016/j.jare.2024.08.038] [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: 06/15/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Host-microbe interactions are important to human health and ecosystems globally, so elucidating the complex host-microbe interactions and associated protein expressions drives the need to develop sensitive and accurate biochemical techniques. Current proteomics techniques reveal information from the point of view of either the host or microbe, but do not provide data on the corresponding partner. Moreover, it remains challenging to simultaneously study host-microbe proteomes that reflect the direct competition between host and microbe. This raises the need to develop a dual-species proteomics method for host-microbe interactions. OBJECTIVES We aim to establish a forward + reverse Stable Isotope Labeling with Amino acids in Cell culture (SILAC) proteomics approach to simultaneously label and quantify newly-expressed proteins of host and microbe without physical isolation, for investigating mechanisms in direct host-microbe interactions. METHODS Using Caenorhabditis elegans-Pseudomonas aeruginosa infection model as proof-of-concept, we employed SILAC proteomics and molecular pathway analysis to characterize the differentially-expressed microbial and host proteins. We then used molecular docking and chemical characterization to identify chemical inhibitors that intercept host-microbe interactions and eliminate microbial infection. RESULTS Based on our proteomics results, we studied the iron competition between pathogen iron scavenger and host iron uptake protein, where P. aeruginosa upregulated pyoverdine synthesis protein (PvdA) (fold-change of 5.2313) and secreted pyoverdine, and C. elegans expressed ferritin (FTN-2) (fold-change of 3.4057). Targeted intervention of iron competition was achieved using Galangin, a ginger-derived phytochemical that inhibited pyoverdine production and biofilm formation in P. aeruginosa. The Galangin-ciprofloxacin combinatorial therapy could eliminate P. aeruginosa biofilms in a fish wound infection model, and enabled animal survival. CONCLUSION Our work provides a novel SILAC-based proteomics method that can simultaneously evaluate host and microbe proteomes, with future applications in higher host organisms and other microbial species. It also provides insights into the mechanisms dictating host-microbe interactions, offering novel strategies for anti-infective therapy.
Collapse
Affiliation(s)
- Yang Sylvia Liu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region
| | - Chengqian Zhang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong Special Administrative Region; City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, China.
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
45
|
Fernandes ÂR, Rodrigues AG, Cobrado L. Effect of prolonged exposure to disinfectants in the antimicrobial resistance profile of relevant micro-organisms: a systematic review. J Hosp Infect 2024; 151:45-59. [PMID: 38740303 DOI: 10.1016/j.jhin.2024.04.017] [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: 03/01/2024] [Revised: 04/08/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Antimicrobial resistance (AMR) constitutes a major global health threat, to a very large extent due to the inadequate use of antibiotics. Additionally, the misuse of disinfectants can also trigger the selection of resistant clones, where micro-organisms develop an adaptative response and progress to resistance mechanisms. Cross-resistance may occur when a biocide's selective pressure induces antimicrobial resistance. AIM To acknowledge the potential relationship between repeated and/or prolonged exposure to disinfectants and antimicrobial resistance profile adjustment. METHODS This systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Studies published until December 2023 that were related to the interaction between disinfectants and antimicrobials were included. Further selection was based on the methodology of exposure. FINDINGS Selected studies included testing about 'exposure to sublethal concentrations' for seventeen disinfectants. The mechanism of action for the majority of the disinfectants involved interactions with the cell membrane. Chlorhexidine was the most studied disinfectant. CONCLUSION Adaptation phenomena related to disinfectant exposure were documented and development of cross-resistance to antimicrobials was verified for several species, including Streptococcus spp., Klebsiella pneumoniae, Acinetobacter baumannii, Staphylococcus aureus, Pseudomonas aeruginosa, and Candida spp. Changes associated with disinfectant exposure also influenced biofilm formation, colony morphology, and efflux pump activity - three relevant determinants of loss of antibiotic efficacy.
Collapse
Affiliation(s)
- Â R Fernandes
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - A G Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal; Centre for Health Technology and Services Research/Rede de Investigação em Saúde (CINTESIS@RISE), Faculty of Medicine, University of Porto, Porto, Portugal; Burn Unit, Department of Plastic and Reconstructive Surgery, University Hospital Centre of São João, Porto, Portugal
| | - L Cobrado
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal; Centre for Health Technology and Services Research/Rede de Investigação em Saúde (CINTESIS@RISE), Faculty of Medicine, University of Porto, Porto, Portugal; Burn Unit, Department of Plastic and Reconstructive Surgery, University Hospital Centre of São João, Porto, Portugal
| |
Collapse
|
46
|
Conte R, Valentino A, De Luca I, Soares Pontes G, Calarco A, Cerruti P. Thermo-Responsive Hydrogel Containing Microfluidic Chitosan Nanoparticles Loaded with Opuntia ficus-indica Extract for Periodontitis Treatment. Int J Mol Sci 2024; 25:9374. [PMID: 39273327 PMCID: PMC11395269 DOI: 10.3390/ijms25179374] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease resulting from the dysbiosis of periodontal bacteria and the host's immune response, leading to tissue degradation and sustained inflammation. Traditional treatments, such as mechanical debridement and antimicrobial agents, often fail to fully eradicate pathogenic bacteria, especially in deep periodontal pockets. Consequently, the need for novel therapeutic approaches has increased the interest in bioactive natural extracts, such as that of Opuntia ficus-indica, known for its anti-inflammatory, antioxidant, and antimicrobial properties. This study investigates the encapsulation of Opuntia ficus-indica extract in OFI-loaded chitosan nanoparticles (OFI-NPs) via ionotropic gelation using a microfluidic system, allowing precise control over nanoparticle characteristics and enhancing protection against enzymatic degradation. To achieve localized and sustained release in periodontal pockets, a thermo-responsive hydrogel comprising hyaluronic acid and Pluronic F127 (OFI@tgels) was developed. The transition of OFI@tgels from a solution at low temperatures to a solid at body temperature enables prolonged drug release at inflammation sites. The in vitro application of the optimized formulation eradicated biofilms of S. mutans, P. aeruginosa (PAO1), and P. gingivalis over 36 h and disrupted extracellular polymeric substance formation. Additionally, OFI@tgel modulated immune responses by inhibiting M1 macrophage polarization and promoting a shift to the M2 phenotype. These findings suggest that OFI@tgel is a promising alternative treatment for periodontitis, effectively reducing biofilm formation and modulating the immune response.
Collapse
Affiliation(s)
- Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Anna Valentino
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Ilenia De Luca
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Gemilson Soares Pontes
- Laboratory of Virology and Immunology, National Institute of Amazonian Research (INPA), Manaus 69067-375, AM, Brazil
- Post-Graduate Program in Basic and Applied Immunology, Institute of Biological Science, Federal University of Amazonas, Manaus 69077-000, AM, Brazil
| | - Anna Calarco
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Pierfrancesco Cerruti
- Institute for Polymers, Composites, and Biomaterials (IPCB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| |
Collapse
|
47
|
Gong Y, Wang H, Sun J. AMP-Mimetic Antimicrobial Polymer-Involved Synergic Therapy with Various Coagents for Improved Efficiency. Biomacromolecules 2024; 25:4619-4638. [PMID: 38717069 DOI: 10.1021/acs.biomac.3c01458] [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: 08/13/2024]
Abstract
The misuse of antibiotics contributes to the emergence of multidrug-resistant (MDR) bacteria. Infections caused by MDR bacteria are rapidly evolving into a significant threat to global healthcare due to the lack of effective and safe treatments. Antimicrobial peptides (AMPs) with broad-spectrum antibacterial activity kill bacteria generally through a membrane disruption mechanism; hence, they tend not to induce resistance readily. However, AMPs exhibit disadvantages, such as high cost and susceptibility to proteolytic degradation, which limit their clinical application. AMP-mimetic antimicrobial polymers, with low cost, stability to proteolysis, broad-spectrum antimicrobial activity, negligible antimicrobial resistance, and rapid bactericidal effect, have received extensive attention as a new type of antibacterial drugs. Lately, AMP-mimetic polymer-involved synergic therapy provides a superior alternative to combat MDR bacteria by distinct mechanisms. In this Review, we summarize the AMP-mimetic antimicrobial polymers involved in synergic therapy, particularly focusing on the different combinations between the polymers with commercially available antimicrobials, organic small molecule photosensitizers, inorganic nanomaterials, and nitric oxide.
Collapse
Affiliation(s)
- Yiyu Gong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, P. R. China
| | - Hepeng Wang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P. R. China
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, P. R. China
| |
Collapse
|
48
|
Conway J, Delanois RE, Mont MA, Stavrakis A, McPherson E, Stolarski E, Incavo S, Oakes D, Salvagno R, Adams JS, Kisch-Hancock A, Tenorio E, Leighton A, Ryser S, Kauvar LM, Bernthal NM. Phase 1 study of the pharmacokinetics and clinical proof-of-concept activity of a biofilm-disrupting human monoclonal antibody in patients with chronic prosthetic joint infection of the knee or hip. Antimicrob Agents Chemother 2024; 68:e0065524. [PMID: 39012102 PMCID: PMC11304715 DOI: 10.1128/aac.00655-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Abstract
We report the results of a first-in-human phase 1 clinical study to evaluate TRL1068, a native human monoclonal antibody that disrupts bacterial biofilms with broad-spectrum activity against both Gram-positive and Gram-negative species. The study population consisted of patients with chronic periprosthetic joint infections (PJIs) of the knee or hip, including both monomicrobial and polymicrobial infections, that are highly resistant to antibiotics due to biofilm formation. TRL1068 was administered via a single pre-surgical intravenous infusion in three sequentially ascending dose groups (6, 15, and 30 mg/kg). Concomitant perioperative antibiotics were pathogen-targeted as prescribed by the treating physician. In this double-blinded study, 4 patients were randomized to receive placebo and 11 patients to receive TRL1068 on day 1, as well as targeted antibiotics for 7 days prior to the scheduled removal of the infected implant and placement of an antibiotic-eluting spacer as the first stage of the standard of care two-stage exchange arthroplasty. No adverse events attributable to TRL1068 were reported. TRL1068 serum half-life was 15-18 days. At day 8, the concentration in synovial fluid was approximately 60% of the blood level and thus at least 15-fold above the threshold for biofilm-disrupting activity in vitro. Explanted prostheses were sonicated to release adherent bacteria for culture, with elimination of the implant bacteria observed in 3 of the 11 patients who received TRL1068, which compares favorably to prior PJI treatments. None of the patients who received TRL1068 had a relapse of the original infection by the end of the study (day 169). CLINICAL TRIALS This study is registered with ClinicalTrials.gov as NCT04763759.
Collapse
Affiliation(s)
- Janet Conway
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, Maryland
| | - Ronald E. Delanois
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, Maryland
| | - Michael A. Mont
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, Maryland
| | | | - Edward McPherson
- University of California Los Angeles, Los Angeles, California, USA
| | | | | | - Daniel Oakes
- University of Southern California, Los Angeles, California, USA
| | | | - John S. Adams
- University of California Los Angeles, Los Angeles, California, USA
| | | | - Edgar Tenorio
- Trellis Bioscience, Inc., Redwood City, California, USA
| | | | - Stefan Ryser
- Trellis Bioscience, Inc., Redwood City, California, USA
| | | | | |
Collapse
|
49
|
Alves F, Nakada PJT, Marques MJDAM, Rea LDC, Cortez AA, Pellegrini VDOA, Polikarpov I, Kurachi C. Complete photodynamic inactivation of Pseudomonas aeruginosa biofilm with use of potassium iodide and its comparison with enzymatic pretreatment. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 257:112974. [PMID: 38964021 DOI: 10.1016/j.jphotobiol.2024.112974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/06/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Pseudomonas aeruginosa, a gram-negative bacterium, accounts for 7% of all hospital-acquired infections. Despite advances in medicine and antibiotic therapy, P. aeruginosa infection still results in high mortality rates of up to 62% in certain patient groups. This bacteria is also known to form biofilms, that are 10 to 1000 times more resistant to antibiotics compared to their free-floating counterparts. Photodynamic Inactivation (PDI) has been proved to be an effective antimicrobial technique for microbial control. This method involves the incubation of the pathogen with a photosensitizer (PS), then, a light at appropriated wavelength is applied, leading to the production of reactive oxygen species that are toxic to the microbial cells. Studies have focused on strategies to enhance the PDI efficacy, such as a pre-treatment with enzymes to degrade the biofilm matrix and/or an addition of inorganic salts to the PS. The aim of the present study is to evaluate the effectiveness of PDI against P. aeruginosa biofilm in association with the application of the enzymes prior to PDI (enzymatic pre-treatment) or the addition of potassium iodide (KI) to the photosensitizer solution, to increase the inactivation effectiveness of the treatment. First, a range of enzymes and PSs were tested, and the best protocols for combined treatments were selected. The results showed that the use of enzymes as a pre-treatment was effective to reduce the total biomass, however, when associated with PDI, mild bacterial reductions were obtained. Then, the use of KI in association with the PS was evaluated and the results showed that, PDI mediated by methylene blue (MB) in the presence of KI was able to completely eradicate the biofilm. However, when the PDI was performed with curcumin and KI, no additive reduction was observed. In conclusion, out of all strategies evaluated in the present study, the most promising strategy to improve PDI against P. aeruginosa biofilm was the use of KI in association with MB, resulting in eradication with 108 log bacterial inactivation.
Collapse
Affiliation(s)
- Fernanda Alves
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Paulo Júnior Tadayoshi Nakada
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Maria Júlia de Arruda Mazzotti Marques
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Leonardo da Cruz Rea
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Anelyse Abreu Cortez
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Vanessa de Oliveira Arnoldi Pellegrini
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Igor Polikarpov
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil
| | - Cristina Kurachi
- São Carlos Institute of Physics (IFSC), University of São Paulo (USP), Brazil, Av. Trabalhador São-carlense, 400 - Centro, CEP 13560-970 São Carlos, SP, Brazil.
| |
Collapse
|
50
|
Buckner E, Buckingham-Meyer K, Miller LA, Parker AE, Jones CJ, Goeres DM. Coupon position does not affect Pseudomonas aeruginosa and Staphylococcus aureus biofilm densities in the CDC biofilm reactor. J Microbiol Methods 2024; 223:106960. [PMID: 38788980 DOI: 10.1016/j.mimet.2024.106960] [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: 04/05/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
The CDC Biofilm Reactor method is the standard biofilm growth protocol for the validation of US Environmental Protection Agency biofilm label claims. However, no studies have determined the effect of coupon orientation within the reactor on biofilm growth. If positional effects have a statistically significant impact on biofilm density, they should be accounted for in the experimental design. Here, we isolate and quantify biofilms from each possible coupon surface in the reactor to quantitatively determine the positional effects in the CDC Biofilm Reactor. The results showed no statistically significant differences in viable cell density across different orientations and vertical positions in the reactor. Pseudomonas aeruginosa log densities were statistically equivalent among all coupon heights and orientations. While the Staphylococcus aureus cell growth showed no statistically significant differences, the densities were not statistically equivalent among all coupon heights and orientations due to the variability in the data. Structural differences were observed between biofilms on the high-shear baffle side of the reactor compared to the lower shear glass side of the reactor. Further studies are required to determine whether biofilm susceptibility to antimicrobials differs based on structural differences attributed to orientation.
Collapse
Affiliation(s)
- Elizabeth Buckner
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, United States of America.
| | - Kelli Buckingham-Meyer
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, United States of America.
| | - Lindsey A Miller
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, United States of America.
| | - Albert E Parker
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, United States of America.
| | - Christopher J Jones
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, United States of America.
| | - Darla M Goeres
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, United States of America.
| |
Collapse
|