Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,704)

Search Parameters:
Keywords = biofilm infection

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 250 KB  
Article
Improved Microbiological Diagnosis of Bone and Joint Infections Using Mechanical Bead-Milling Extraction of Bone Specimens with the Ultra-Turrax® System
by Maxime Brunaud, Adeline Boutet-Dubois, Alix Pantel, Florian Salipante, Rémy Coulomb, Albert Sotto, Jean-Philippe Lavigne and Nicolas Cellier
Diagnostics 2026, 16(2), 309; https://doi.org/10.3390/diagnostics16020309 (registering DOI) - 18 Jan 2026
Abstract
Background: Accurate microbiological diagnosis of bone and joint infections (BJIs) is frequently hampered by low bacterial load, biofilm formation, and suboptimal tissue processing. This study evaluated the diagnostic performance of mechanical bead-milling using the Ultra-Turrax® Tube Drive system compared with standard [...] Read more.
Background: Accurate microbiological diagnosis of bone and joint infections (BJIs) is frequently hampered by low bacterial load, biofilm formation, and suboptimal tissue processing. This study evaluated the diagnostic performance of mechanical bead-milling using the Ultra-Turrax® Tube Drive system compared with standard vortex homogenization. Methods: In a prospective cohort of 116 patients undergoing surgery for suspected BJIs, 540 intraoperative samples were processed using both methods. Culture and 16S rRNA PCR results were analyzed using classical and Bayesian statistical approaches. Diagnostic performance was assessed globally and across specimen types and anatomical sites. Results: Ultra-Turrax® significantly improved sensitivity across all sample types (87.1% vs. 75.2%, p < 0.0001), while maintaining comparable specificity (>99%). Culture positivity increased by 17%, with the greatest gains observed in bone samples and hip prosthesis infections. Quantitative cultures demonstrated a 1.5–2 log10 CFU/mL increase in bacterial yield. In culture-negative specimens, 16S rRNA PCR detection doubled with Ultra-Turrax® processing (26% vs. 13%, p = 0.04). No increase in contamination was observed. Time to positivity was similar between methods, although Ultra-Turrax® provided earlier results in 17% of cases. Bayesian modeling confirmed superior sensitivity (posterior probability > 0.995). Conclusions: Ultra-Turrax® bead-milling markedly enhances microbiological detection in BJIs, particularly in low-biomass and bone-derived specimens. Its simplicity, reproducibility, and compatibility with routine workflows support its integration into diagnostic pathways. This pre-analytical optimization may improve etiological identification and guide more targeted antimicrobial therapy. Full article
16 pages, 2452 KB  
Article
Fusobacterium nucleatum Enhances Intestinal Adaptation of Vibrio cholerae via Interspecies Biofilm Formation
by Guozhong Chen, Jiamin Chen, Xiangfeng Wang, Dingming Guo and Zhi Liu
Microorganisms 2026, 14(1), 211; https://doi.org/10.3390/microorganisms14010211 (registering DOI) - 16 Jan 2026
Viewed by 27
Abstract
Biofilm formation represents a key survival strategy employed by Vibrio cholerae to adapt to the complex intestinal environment of the host. While most previous studies on V. cholerae biofilms have focused on genetic regulation and monospecies cultures, its ability to form dual-species biofilms [...] Read more.
Biofilm formation represents a key survival strategy employed by Vibrio cholerae to adapt to the complex intestinal environment of the host. While most previous studies on V. cholerae biofilms have focused on genetic regulation and monospecies cultures, its ability to form dual-species biofilms with other intestinal pathogens is still poorly understood. In this study, using samples from both cholera patients and healthy individuals, Fusobacterium nucleatum was identified as a bacterium capable of co-aggregating with V. cholerae. Untargeted metabolomic analysis revealed that F. nucleatum-derived metabolites, specifically 6-hypoxanthine, enhance biofilm formation in V. cholerae. Further validation confirmed that these F. nucleatum-derived metabolites upregulate the biofilm-associated regulatory gene vpsT. In an adult mouse model, co-infection with F. nucleatum and V. cholerae significantly enhanced the intestinal adaptability of V. cholerae compared to infection with V. cholerae alone. Together, these findings elucidate the mechanism enabling the co-infection of F. nucleatum and V. cholerae in the host intestine, thereby shedding new light on how other pathogenic bacteria can assist in V. cholerae infection. Full article
(This article belongs to the Section Biofilm)
Show Figures

Figure 1

12 pages, 1699 KB  
Article
Non-C. albicans Candida Species Develop Clinically Relevant Biofilms on Stainless Steel Under Respiratory Tract-Mimicking Conditions
by Natalia Bagon, Vlaudimir Marques, Deisiany Ferreira and Melyssa Negri
Life 2026, 16(1), 148; https://doi.org/10.3390/life16010148 - 16 Jan 2026
Viewed by 52
Abstract
Biofilm formation by non-C. albicans Candida (NAC) species is a major factor in device-associated infections, yet few studies have examined their development under physiologically relevant conditions. This study evaluated the biofilm-forming capacity of Candida tropicalis, Candida parapsilosis sensu stricto and Candida [...] Read more.
Biofilm formation by non-C. albicans Candida (NAC) species is a major factor in device-associated infections, yet few studies have examined their development under physiologically relevant conditions. This study evaluated the biofilm-forming capacity of Candida tropicalis, Candida parapsilosis sensu stricto and Candida albicans on stainless steel surfaces in the presence of artificial saliva, simulating the respiratory tract environment of tracheostomized patients. Standardized inocula were incubated for 24 h, and biofilms were assessed through quantification of viable cells, biomass, biofilm matrix production and structural characterization by scanning electron microscopy (SEM). C. tropicalis produced the most robust biofilms compared to C. albicans and C. parapsilosis stricto sensu isolates, with significantly higher biomass and biofilm matrix (p < 0.001). C. parapsilosis sensu stricto developed less dense yet structurally defined biofilm networks. SEM confirmed mature and compact biofilm architecture, especially in C. tropicalis. These results demonstrate the strong intrinsic biofilm-forming ability of NAC species on stainless steel under host-like conditions, reinforcing their capacity to persist on medical surfaces and their relevance as independent contributors to biofilm-related contamination and infection. Full article
(This article belongs to the Section Microbiology)
Show Figures

Figure 1

17 pages, 539 KB  
Review
Phage Therapy at the Crossroads Between Clinical Promise and Regulatory Challenge
by Anna Gallina, Matteo Gallina, Andrea Cona, Patrizio Vitulo, Alessandra Mularoni and Alessio Provenzani
Pharmaceuticals 2026, 19(1), 162; https://doi.org/10.3390/ph19010162 - 16 Jan 2026
Viewed by 157
Abstract
Bacteriophage (phage) therapy, including monophage preparations, phage cocktails, engineered phages, and phage-derived enzymes, has re-emerged as a potential option for difficult-to-treat and biofilm-associated infections in the context of rising antimicrobial resistance. Recent scientific and regulatory developments, such as the 2024 World Health Organization [...] Read more.
Bacteriophage (phage) therapy, including monophage preparations, phage cocktails, engineered phages, and phage-derived enzymes, has re-emerged as a potential option for difficult-to-treat and biofilm-associated infections in the context of rising antimicrobial resistance. Recent scientific and regulatory developments, such as the 2024 World Health Organization Bacterial Priority Pathogens List and the introduction of the European Pharmacopoeia general chapter 5.31 on phage therapy medicinal products, highlight the growing interest in establishing quality, safety, and governance standards for clinical implementation. This narrative review provides an overview of current clinical applications of phage therapy, drawing on published case reports, case series, early-phase clinical studies, and regulatory experiences across different healthcare settings. Clinical use has been reported in respiratory, urinary tract, musculoskeletal, cardiovascular, and device-associated infections, particularly in cases involving multidrug-resistant pathogens, often in combination with antibiotics. At the same time, the biological characteristics of phages, such as strain specificity, adaptive composition of phage cocktails, and the need for individualized formulations, pose significant regulatory and translational challenges. Access to phage therapy currently relies on heterogeneous regulatory mechanisms, including compassionate use programmes, magistral preparations, named-patient pathways, and other national frameworks. Overall, phage therapy represents a promising strategy for selected infections, but its broader clinical adoption will depend on harmonized regulatory approaches, robust quality standards, and the generation of stronger clinical evidence to support safe and scalable use. Full article
Show Figures

Figure 1

39 pages, 2612 KB  
Review
Marine Bacteria as a Source of Antibiotics Against Staphylococcus aureus: Natural Compounds, Mechanisms of Action, and Discovery Strategies
by Céphas Xuma, Alexandre Bourles, Julien Colot, Linda Guentas and Mariko Matsui
Mar. Drugs 2026, 24(1), 44; https://doi.org/10.3390/md24010044 - 15 Jan 2026
Viewed by 300
Abstract
Staphylococcus aureus is a major opportunistic pathogen responsible for a wide spectrum of human infections, including severe and difficult-to-treat cases. The emergence of multidrug-resistant strains limits the efficacy of conventional antibiotic therapies and poses a significant global public health challenge. In this context, [...] Read more.
Staphylococcus aureus is a major opportunistic pathogen responsible for a wide spectrum of human infections, including severe and difficult-to-treat cases. The emergence of multidrug-resistant strains limits the efficacy of conventional antibiotic therapies and poses a significant global public health challenge. In this context, the search for novel antibiotics has intensified, with increasing interest in marine resources, an ecosystem still largely underexplored. Marine bacteria produce a vast array of secondary metabolites with unique structures and potentially novel modes of antibacterial action. Several compounds isolated from marine bacterial strains have demonstrated promising activity against multidrug-resistant S. aureus, including antivirulence effects such as biofilm formation and Quorum-Sensing inhibition. This review explores the potential of marine bacteria as a source of new antibiotics against S. aureus, discusses both classical and advanced strategies for the discovery of bioactive molecules, and highlights the scientific and technological challenges involved in translating these findings into clinical applications. Full article
(This article belongs to the Section Marine Pharmacology)
Show Figures

Figure 1

10 pages, 447 KB  
Article
An Unexpected Inverse Relationship Between Biofilm Formation and Antibiotic Resistance in Stenotrophomonas maltophilia
by Arianna Pompilio and Giovanni Di Bonaventura
Antibiotics 2026, 15(1), 85; https://doi.org/10.3390/antibiotics15010085 - 15 Jan 2026
Viewed by 94
Abstract
Background/Objectives: Stenotrophomonas maltophilia is an opportunistic pathogen causing severe infections, particularly in patients with cystic fibrosis (CF). Its intrinsic multidrug resistance and biofilm-forming capacity complicate treatment. Although biofilms are generally associated with antimicrobial tolerance, the relationship between biofilm formation and planktonic antibiotic [...] Read more.
Background/Objectives: Stenotrophomonas maltophilia is an opportunistic pathogen causing severe infections, particularly in patients with cystic fibrosis (CF). Its intrinsic multidrug resistance and biofilm-forming capacity complicate treatment. Although biofilms are generally associated with antimicrobial tolerance, the relationship between biofilm formation and planktonic antibiotic resistance in S. maltophilia remains poorly understood. This study investigated the association between antibiotic resistance profiles and biofilm production in clinical isolates from CF and non-CF patients. Methods: A total of 86 clinical isolates (40 from CF airways and 46 from non-CF patients) were analyzed. Susceptibility to seven antibiotics was assessed by disk diffusion, and multidrug resistance profiles were defined using standard criteria. Biofilm formation was quantified after 24 h using a crystal violet microtiter plate assay and categorized by using a semiquantitative scale. Results: High resistance rates were observed, particularly to meropenem (87.2%), ciprofloxacin (80.2%), and rifampicin (72.1%). CF isolates exhibited significantly higher resistance to piperacillin/tazobactam and a greater prevalence of multidrug resistance. Biofilm formation was detected in 94.2% of isolates, with strong or powerful producers predominating. However, CF isolates formed significantly less biofilm than non-CF isolates. Notably, resistance to piperacillin/tazobactam and meropenem was associated with reduced biofilm biomass and a lower proportion of high biofilm producers. Across all isolates, an inverse correlation was observed between the number of antibiotic resistances and biofilm biomass. These trends persisted after stratification by clinical origin, although some comparisons did not reach statistical significance. Conclusions: This study reveals an unexpected inverse relationship between planktonic antibiotic resistance and biofilm-forming capacity in S. maltophilia. Enhanced biofilm production may represent an alternative persistence strategy in more antibiotic-susceptible strains, with important implications for infection management and therapeutic failure. Full article
Show Figures

Graphical abstract

30 pages, 4170 KB  
Article
EruA, a Regulator of Adherent-Invasive E. coli, Enhances Bacterial Pathogenicity by Promoting Adhesion to Epithelial Cells and Survival Within Macrophages
by Zeyan Xu, Chuyu Qin, Ruohan Zhang, Mengting Wu, Anqi Cui, Wei Chen, Lu Chen, Daqing Gao and Ruihua Shi
Biomolecules 2026, 16(1), 152; https://doi.org/10.3390/biom16010152 - 14 Jan 2026
Viewed by 165
Abstract
Adherent-invasive E. coli (AIEC) is closely related to inflammatory bowel disease (IBD). However, its pathogenic mechanism has not yet been fully elucidated. Using a BLASTP search, we discovered that the amino acid sequence of a putative protein (UFP37798.1) in the AIEC LF82 strain [...] Read more.
Adherent-invasive E. coli (AIEC) is closely related to inflammatory bowel disease (IBD). However, its pathogenic mechanism has not yet been fully elucidated. Using a BLASTP search, we discovered that the amino acid sequence of a putative protein (UFP37798.1) in the AIEC LF82 strain is highly homologous to some regulators in the SlyA family. We named it EruA. We displayed the secondary structures of EruA using bioinformatics, overexpressed the His6-tagged EruA protein using SDS-PAGE, and dissected the genetic organization of the eruA chromosomal region using 5′RACE. We constructed an eruA deletion mutant (ΔeruA) and a complementary strain (CΔeruA) of the LF82 strain. The transcriptomes of wild-type (WT) and ΔeruA bacteria were compared using RNA sequencing and qRT-PCR, thereby identifying 32 differentially expressed genes (DEGs). Based on YASARA software and EMSA analysis, EruA directly binds to the consensus sequences (PfimA and PtnaB) in the promoter region of the fimA and tnaB genes from these DEGs. By using a super-resolution confocal microscope (SCM), counting CFUs of colonies on plates, indole quantification, and crystal violet staining of biofilms adhered to tubes or 96-well plates, we found that EruA activates the fimA to promote bacterial adhesion to intestinal epithelial cells and activates the tnaB to enhance bacterial indole production and biofilm formation. Moreover, EruA helps AIEC resist environmental stress and enhances bacterial survival within macrophages as well as loading in mouse tissues. Notably, EruA promotes AIEC colonization in the colons of mice and exacerbates intestinal inflammation caused by bacterial infection in mice with DSS-induced inflammatory colitis, manifested by weight loss, colon length shortening, and pathological changes in colon tissues. Therefore, EruA plays a key role in the pathogenicity of AIEC. Full article
(This article belongs to the Special Issue Recent Advances in Molecular Genetics of Bacteria)
Show Figures

Figure 1

14 pages, 1372 KB  
Article
Carvacrol Reduces Virulence Traits in Meyerozyma guilliermondii and Candida dubliniensis and Enhances Galleria mellonella Survival During Candidozyma auris Infection
by Andrea Giammarino, Laura Verdolini, Javier Mussin, Giulia Radocchia, Florencia Rojas, Gustavo Giusiano and Letizia Angiolella
Microorganisms 2026, 14(1), 188; https://doi.org/10.3390/microorganisms14010188 - 14 Jan 2026
Viewed by 133
Abstract
Background: Antifungal resistance among Candida species and related genera, coupled with the lack of new drugs, poses a significant threat to public health. Several studies have demonstrated a relationship between virulence factors and resistance. Current objectives include identifying new targets and searching for [...] Read more.
Background: Antifungal resistance among Candida species and related genera, coupled with the lack of new drugs, poses a significant threat to public health. Several studies have demonstrated a relationship between virulence factors and resistance. Current objectives include identifying new targets and searching for new natural molecules. Carvacrol, a natural phenolic compound, has been shown to have antimicrobial properties; however, its impact on the virulence of species other than Candida albicans and related yeast genera remains underexplored. Methods: The antifungal activity of carvacrol was evaluated against clinical isolates of Candidozyma auris, Meyerozyma guilliermondii, and Candida dubliniensis, as well as its effect on adhesion, hydrophobicity, biofilm formation and osmotic stress tolerance. In vivo activity was assessed using the Galleria mellonella infection model at MIC concentrations. Results: Carvacrol inhibited adherence and significantly reduced both early and preformed biofilms in M. guilliermondii and C. dubliniensis. In C. auris, the compound produced a modest reduction in biofilm activity but significantly enhanced larval survival in the in vivo model (~20%, p < 0.01). Carvacrol also induced increased tolerance of C. auris to osmotic stress, suggesting activation of adaptive pathways. Conclusions: Carvacrol exhibits species-specific effects, acting as an antivirulence modulator in M. guilliermondii and C. dubliniensis and attenuating virulence in vivo in C. auris. These findings support the potential of carvacrol as an adjuvant antifungal strategy, particularly against C. auris, and highlight the relevance of targeting virulence traits to reduce selective pressure and limit antifungal resistance. Full article
(This article belongs to the Special Issue Research on Fungal Pathogen Candida spp. and Alternative Therapy)
Show Figures

Figure 1

19 pages, 1138 KB  
Review
Antifungal Activity of Natural Thymol: Advances on Molecular Mechanisms and Therapeutic Potential
by Chun Chen, Lu Liu, Shusheng Tang, Daowen Li and Chongshan Dai
Biomolecules 2026, 16(1), 149; https://doi.org/10.3390/biom16010149 - 14 Jan 2026
Viewed by 100
Abstract
Currently, the increased incidence of invasive fungal infections globally is posing a significant challenge to public health. Due to drug resistance issues, the clinical efficacy of existing antifungal drugs is seriously insufficient, while new drug development progresses slowly. Consequently, there is an urgent [...] Read more.
Currently, the increased incidence of invasive fungal infections globally is posing a significant challenge to public health. Due to drug resistance issues, the clinical efficacy of existing antifungal drugs is seriously insufficient, while new drug development progresses slowly. Consequently, there is an urgent need to discover and develop novel antifungal therapeutics. Natural products have the characteristics of wide sources and few adverse reactions and are one of the sources for developing antifungal drugs. Numerous studies have shown that many compounds isolated from plants and traditional Chinese medicine have antifungal activity and diverse antifungal mechanisms. Thymol, a monoterpene phenol compound from thyme (Lamiaceae), has multiple biological functions such as antibacterial, antioxidant, and anti-inflammatory. Recent research has found that thymol has strong antifungal activity, and its molecular mechanisms involve cell membrane rupture, interference with cell wall synthesis, disruption of mitochondrial function and energy metabolism, inhibition of biofilm, inhibition of virulence factor expression, inhibition of key enzymes, and induction of cell apoptosis. This review aimed to summarize the antifungal activity of thymol and the underlying molecular mechanisms, safety, and potential clinical applications. Emerging technologies in thymol delivery systems and future research directions are also discussed. The comprehensive analysis aims to provide a detailed understanding of fungal infections and the role of thymol in antifungal treatment, offering insights for further research and clinical practice. Full article
(This article belongs to the Topic Recent Advances in Veterinary Pharmacology and Toxicology)
Show Figures

Figure 1

20 pages, 14008 KB  
Article
The Antimicrobial Peptide CRAMP-34 Eradicates Escherichia coli Biofilms by Interfering with the kduD-Dependent Network
by Hongzao Yang, Jing Xiong, Sisi Su, Zhuo Yang, Wu Yang, Lianci Peng, Suhui Zhang, Jinjie Qiu, Yuzhang He and Hongwei Chen
Antibiotics 2026, 15(1), 83; https://doi.org/10.3390/antibiotics15010083 - 14 Jan 2026
Viewed by 163
Abstract
Background/Objectives: Bacterial biofilms formed by Escherichia coli pose a significant challenge in veterinary medicine due to their intrinsic resistance to antibiotics. Antimicrobial peptides (AMPs) represent a promising alternative. AMPs exert their bactericidal activity by binding to negatively charged phospholipids in bacterial membranes [...] Read more.
Background/Objectives: Bacterial biofilms formed by Escherichia coli pose a significant challenge in veterinary medicine due to their intrinsic resistance to antibiotics. Antimicrobial peptides (AMPs) represent a promising alternative. AMPs exert their bactericidal activity by binding to negatively charged phospholipids in bacterial membranes via electrostatic interactions, leading to membrane disruption and rapid cell lysis. Methods: In vitro assays including MIC determination, biofilm eradication testing (crystal violet, colony counts, and CLSM), swimming motility, and EPS quantification were performed. CRISPR/Cas9 was used to construct and complement a kduD mutant. A transposon mutagenesis library was screened for biofilm-defective mutants. In an in vivo murine excisional wound infection model treated with the mouse cathelicidin-related antimicrobial peptide (CRAMP-34), wound closure and bacterial burden were monitored. Gene expression changes were analyzed via RT-qPCR. Results: CRAMP-34 effectively eradicated pre-formed biofilms of a clinically relevant, porcine-origin E. coli strain and promoted wound healing in the murine infection model. We conducted a genome-wide transposon mutagenesis screen, which identified kduD as a critical gene for robust biofilm formation. Functional characterization revealed that kduD deletion drastically impairs flagellar motility and alters exopolysaccharide production, leading to defective biofilm architecture without affecting growth. Notably, the anti-biofilm activity of CRAMP-34 phenocopied aspects of the kduD deletion, including motility inhibition and transcriptional repression of a common set of biofilm-related genes. Conclusions: This research highlights CRAMP-34 as a potent anti-biofilm agent and unveils kduD as a previously unrecognized regulator of E. coli biofilm development, which is also targeted by CRAMP-34. Full article
(This article belongs to the Special Issue Antimicrobial Resistance in Biofilm-Associated Infections)
Show Figures

Figure 1

18 pages, 1961 KB  
Article
Multitarget Mechanisms of (‒)‒Epigallocatechin-3-Gallate Against MRSA: From SraP L-Lectin Targeting to Synergistic Antibiotic Effects
by Ping Zheng, Peihua Zhang, Yuan Li, Jinzhao Long, Fang Liu and Haiyan Yang
Pathogens 2026, 15(1), 90; https://doi.org/10.3390/pathogens15010090 - 13 Jan 2026
Viewed by 110
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), a major global public health threat due to its broad resistance, urgently requires the development of new antibiotic alternatives. (‒)‒Epigallocatechin-3-gallate (EGCG) is considered a natural bioactive compound with anti-MRSA properties. The L-Lectin module of serine-rich adhesin for platelets (SraP) [...] Read more.
Methicillin-resistant Staphylococcus aureus (MRSA), a major global public health threat due to its broad resistance, urgently requires the development of new antibiotic alternatives. (‒)‒Epigallocatechin-3-gallate (EGCG) is considered a natural bioactive compound with anti-MRSA properties. The L-Lectin module of serine-rich adhesin for platelets (SraP) is considered an important target for blocking MRSA-infected hosts. This study aims to investigate the mechanism of action of EGCG against MRSA. Surface plasmon resonance (SPR), cell adhesion and invasion, biofilm formation, checkerboard assays, RNA sequencing (RNA-seq) and quantitative real-time polymerase chain reaction (qRT-PCR) were performed. The results showed that EGCG bound to SraP L Lectin with high affinity and effectively inhibited MRSA colonization. Additionally, EGCG significantly suppressed pyrimidine metabolism and downregulated related genes, thereby potentially inhibiting bacterial growth. It also markedly reduced the expression of multiple genes associated with β-lactam resistance and inhibited biofilm formation. A strong synergistic effect was observed between EGCG and the bactericidal agent ceftriaxone (CRO). When combined with 10 μg/mL EGCG, CRO required 75% less dosage and exhibited a prolonged antimicrobial effect. In conclusion, EGCG exerts anti-MRSA effects through multiple pathways and represents a promising candidate as an alternative therapeutic agent against MRSA infections. Full article
(This article belongs to the Section Bacterial Pathogens)
Show Figures

Figure 1

28 pages, 509 KB  
Review
Mechanisms of Pseudomonas aeruginosa Resilience Against Antibiotic Treatment and Outlooks of Emerging Treatment Strategies
by Angelika Krūmiņa, Indra Zeltiņa, Paula Simsone, Emile Eulitz, Aigars Reinis and Ludmila Vīksna
Medicina 2026, 62(1), 163; https://doi.org/10.3390/medicina62010163 - 13 Jan 2026
Viewed by 161
Abstract
Pseudomonas aeruginosa is a resilient Gram-negative pathogen frequently implicated in healthcare associated infections, particularly among immunocompromised individuals and those with chronic conditions such as cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), or cancer. It is well known for its high resistance to [...] Read more.
Pseudomonas aeruginosa is a resilient Gram-negative pathogen frequently implicated in healthcare associated infections, particularly among immunocompromised individuals and those with chronic conditions such as cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), or cancer. It is well known for its high resistance to antibiotic treatment. This review briefly mentions P. aeruginosa’s resistance mechanisms, biofilm formation, and virulence factors, while primarily focusing on treatment challenges and recent advancements in therapeutic strategies aimed at overcoming resistance. Covered are novel non-antibiotic interventions such as quorum sensing inhibitors, quorum quenching agents, iron chelators, lectin and efflux pump inhibitors, as well as antimicrobial peptides and nanoparticles. Traditional medicine, phytochemicals, and probiotics are also evaluated. Additionally, this review explores the development of a viable vaccine, bacteriophage therapy, lactoferrin-hypothiocyanite combination, and topical use of electrochemical scaffolds. This review emphasizes the need for extensive safety studies and in vivo validation of these emerging non-antibiotic therapeutic strategies to determine their efficacy, pharmacological behavior, and clinical feasibility before they can be translated into practice. Many of these emerging treatments could play a vital role in future combination therapies by enhancing the efficacy of existing antibiotics and countering resistance and virulence mechanisms. Advancing these approaches from laboratory to clinical application remains a major challenge, making the development of approved therapies or vaccines a critical scientific and public health priority. Full article
(This article belongs to the Section Pharmacology)
23 pages, 5049 KB  
Article
Potential of Fermented Food-Derived Lactiplantibacillus Cell-Free Supernatants to Control Staphylococcus aureus Growth and Biofilm Development
by Lena Ilieva, Vesselin Baev, Mariana Marhova, Galina Yahubyan, Elena Apostolova, Mariyana Gozmanova, Velizar Gochev, Tsvetelina Paunova-Krasteva, Tsvetozara Damyanova, Sonya Kostadinova, Miroslava Gocheva and Ivan Iliev
Int. J. Mol. Sci. 2026, 27(2), 760; https://doi.org/10.3390/ijms27020760 - 12 Jan 2026
Viewed by 125
Abstract
Staphylococcus aureus biofilms represent a critical healthcare challenge, driving chronic infections and antimicrobial resistance. This study investigates the anti-staphylococcal efficacy of two Lactiplantibacillus strains isolated from traditional Bulgarian pickled vegetables (turshiya): L. plantarum IZITR_24 and L. paraplantarum IZITR_13. Combining whole genome sequencing (WGS) [...] Read more.
Staphylococcus aureus biofilms represent a critical healthcare challenge, driving chronic infections and antimicrobial resistance. This study investigates the anti-staphylococcal efficacy of two Lactiplantibacillus strains isolated from traditional Bulgarian pickled vegetables (turshiya): L. plantarum IZITR_24 and L. paraplantarum IZITR_13. Combining whole genome sequencing (WGS) with functional assays, we established a robust genotype-to-phenotype framework to characterize their antimicrobial arsenal. Based on WGS, we identified conserved plantaricin (plnJK, plnEF) clusters in both isolates, with IZITR_13 additionally carrying genes for pediocin and enterolysin A—alongside the confirmed absence of virulence factors. Reconstituted lyophilized cell-free supernatants (LCFSs) were evaluated in dose–response microtiter assays to determine the minimum biofilm inhibitory concentration (MBIC) and minimum inhibitory concentration (MIC). Both strains demonstrated clear, dose-dependent inhibitory activity against the S. aureus growth and biofilm formation. Microscopy (SEM/CLSM) confirmed significant biofilm disruption and cell membrane permeabilization. The observed consistency between genome-inferred capacity and phenotypes highlights the strong predictive value of a genome-first screening approach for selecting bacteriocin-producing lactic acid bacteria (LAB). These findings position IZITR_24 and IZITR_13 as promising postbiotic producers with potent antibiofilm activity against S. aureus. By utilizing their stable postbiotic products rather than relying on live colonization, this study proposes a targeted, antibiotic-sparing strategy to combat persistent staphylococcal biofilms. Full article
(This article belongs to the Special Issue Antimicrobial Materials: Molecular Developments and Applications)
Show Figures

Figure 1

12 pages, 266 KB  
Review
Treatment of Periprosthetic Joint Infection After Tumor Megaprosthetic Reconstruction: A Narrative Review
by Wei Wang, Haoran Qiao, Zhiqing Zhao and Taiqiang Yan
Cancers 2026, 18(2), 230; https://doi.org/10.3390/cancers18020230 - 12 Jan 2026
Viewed by 202
Abstract
Purpose: Periprosthetic joint infection (PJI) is a devastating complication following limb salvage surgery with tumor megaprosthetic reconstruction, leading to high morbidity and complex management. Despite advancements in prosthesis design and materials, infection rates are notably higher than in conventional arthroplasty. This narrative review [...] Read more.
Purpose: Periprosthetic joint infection (PJI) is a devastating complication following limb salvage surgery with tumor megaprosthetic reconstruction, leading to high morbidity and complex management. Despite advancements in prosthesis design and materials, infection rates are notably higher than in conventional arthroplasty. This narrative review synthesizes current evidence on the etiology, diagnosis, and management of PJIs in this unique setting. Methods: We conducted narrative review of literature from PubMed and Embase using keywords related to PJIs and tumor megaprostheses, aiming to summarize risk factors, diagnostic criteria, pathogen profiles, and treatment outcomes. Results: Key findings indicate that the risk of PJI is multifactorial, involving patient-related, disease-related, and treatment-related factors. Diagnosis relies on a combination of clinical presentation, serological markers, imaging, and microbiological studies, though established criteria for conventional PJI may require adaptation for tumor cases. Treatment strategies include irrigation and debridement (I&D), debridement, antibiotics, implant retention with modular component exchange (DAIR), one-stage or two-stage revision, and amputation. Success rates vary, and optimal management requires a multidisciplinary, individualized approach. However, two-stage revision is considered the gold standard for chronic PJIs. Conclusions: PJIs after tumor megaprosthetic reconstruction presents distinct challenges. Management requires a multidisciplinary, individualized approach. Future research should focus on validated diagnostic criteria for this population, novel anti-biofilm strategies, and standardized treatment protocols. Full article
14 pages, 576 KB  
Article
Cathelicidin-like Peptide for Resistant Acinetobacter baumannii Control
by Elizabete de Souza Cândido, Danieli Fernanda Buccini, Elizangela de Barros Miranda, Regina Meneses Gonçalves, Amanda Loren de Oliveira Brandão, Valentina Nieto-Marín, Ana Paula Ferreira Leal, Samilla Beatriz Rezende, Marlon Henrique Cardoso and Octavio Luiz Franco
Antibiotics 2026, 15(1), 77; https://doi.org/10.3390/antibiotics15010077 - 12 Jan 2026
Viewed by 144
Abstract
The growing global threat of antimicrobial resistance (AMR), particularly in cutaneous wound infections, represents a significant clinical and economic challenge. Biofilm formation by multidrug-resistant pathogens, such as Acinetobacter baumannii, often complicates healing and leads to therapeutic failure. Antimicrobial peptides (AMPs) are a [...] Read more.
The growing global threat of antimicrobial resistance (AMR), particularly in cutaneous wound infections, represents a significant clinical and economic challenge. Biofilm formation by multidrug-resistant pathogens, such as Acinetobacter baumannii, often complicates healing and leads to therapeutic failure. Antimicrobial peptides (AMPs) are a promising alternative to conventional antibiotics due to their potent membrane-disrupting mechanism of action and lower propensity to induce resistance. Background/Objectives: This study aimed to evaluate the antibacterial, antibiofilm, and in vivo efficacy of four snake venom-derived cathelicidin-like peptides—Btn (15-34) and BotrAMP14 from Bothrops atrox, and Ctn (15-34) and CrotAMP14 from Crotalus durissus—against multidrug-resistant A. baumannii, Escherichia coli, and Pseudomonas aeruginosa clinical isolates from skin infections, with emphasis on A. baumannii, a WHO priority pathogen. Methods: Minimal Inhibitory Concentration (MIC), Minimal Bactericidal Concentration (MBC), and Minimal Biofilm Inhibitory Concentration (MBIC) were determined against A. baumannii, Escherichia coli, and Pseudomonas aeruginosa. Time-kill kinetics, hemolytic activity, and cytotoxicity assays were performed. A murine skin wound infection model was established to evaluate in vivo antibacterial efficacy and safety. Results: MIC/MBC values ranged from 0.78 to 25 µM against planktonic cells. In comparison, MBIC ranged from 1.56 to 12.5 µM against biofilms. BotrAMP14 eradicated A. baumannii within 4 min, while CrotAMP14 achieved bactericidal action in 20 min at 1.56 µM. Both peptides exhibited no hemolytic activity up to 128 µM and low cytotoxicity (IC50 > 128 µM). In vivo, BotrAMP14 and CrotAMP14 demonstrated significant antibacterial activity at 24 h and 48 h post-infection, respectively, surpassing that of meropenem. Conclusions: These findings suggest that BotrAMP14 and CrotAMP14 are promising topical antimicrobial agents for managing multidrug-resistant skin infections and may help address the urgent need for alternative therapies against antibiotic-resistant pathogens. Full article
Show Figures

Graphical abstract

Back to TopTop