[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (757)

Search Parameters:
Keywords = biofilm formation assay

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 525 KiB  
Article
Chemical Profile and Bioactivities of Three Species of Mentha Growing in the Campania Region, Southern Italy
by Rosaria Francolino, Mara Martino, Filomena Nazzaro, Carmina Sirignano, Florinda Fratianni, Francesca Coppola, Laura De Martino, Carmen Formisano and Vincenzo De Feo
Plants 2025, 14(3), 360; https://doi.org/10.3390/plants14030360 - 24 Jan 2025
Viewed by 515
Abstract
The genus Mentha (Lamiaceae), comprising aromatic perennial plants widely distributed in temperate regions, holds significant medicinal and commercial value. This study aimed to investigate the chemical profile and bioactivities of hydroalcoholic extracts from Mentha longifolia (L.) L., Mentha pulegium L., and Mentha spicata [...] Read more.
The genus Mentha (Lamiaceae), comprising aromatic perennial plants widely distributed in temperate regions, holds significant medicinal and commercial value. This study aimed to investigate the chemical profile and bioactivities of hydroalcoholic extracts from Mentha longifolia (L.) L., Mentha pulegium L., and Mentha spicata L. harvested from the Campania region, Southern Italy. Chemical analysis using LC-HRESIMS/MS identified a total of 21 compounds. The extracts, particularly M. pulegium, exhibited notable antioxidant activity, evaluated through DPPH and FRAP assays, probably related to their chemical composition. Both M. pulegium and M. longifolia demonstrated a higher phenolic content, with M. pulegium also containing the highest levels of flavonoids. In addition, the extract’s ability to inhibit biofilm formation was evaluated against several pathogenic strains, including Gram-positive bacteria (Listeria monocytogenes and Staphylococcus aureus) and Gram-negative bacteria (Acinetobacter baumannii, Pseudomonas aeruginosa, and Escherichia coli) using crystal violet and MTT assays. All extracts effectively inhibited biofilm formation in A. baumannii and P. aeruginosa, with M. pulegium also showing moderate activity against the metabolism of L. monocytogenes. The pronounced antibacterial and biofilm-inhibitory properties of M. pulegium highlight its potential for pharmaceutical applications. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Total phenolic and (<b>b</b>) flavonoid contents in <span class="html-italic">M. longifolia</span>, <span class="html-italic">M. pulegium</span>, and <span class="html-italic">M. spicata</span>. GAE = gallic acid equivalents. QE = quercetin equivalents. Values were expressed as mean ± SD from three independent experiments. Different letters within the same graph indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 (two-way ANOVA followed by Tukey’s post hoc test).</p>
Full article ">
14 pages, 1302 KiB  
Article
An In Vitro Study of the Anti-Acne Effects of Scutellaria barbata
by Qiwen Zheng, Xiangji Jin, Trang Thi Minh Nguyen, Se-Jig Park, Gyeong-Seon Yi, Su-Jin Yang and Tae-Hoo Yi
Molecules 2025, 30(3), 515; https://doi.org/10.3390/molecules30030515 - 23 Jan 2025
Viewed by 197
Abstract
Acne is a common skin disease that is closely associated with Cutibacterium acnes (C. acnes) and the inflammatory response it induces. Existing antibiotic treatments are often rendered ineffective due to the development of bacterial resistance, while Scutellaria barbata (SLB) has attracted [...] Read more.
Acne is a common skin disease that is closely associated with Cutibacterium acnes (C. acnes) and the inflammatory response it induces. Existing antibiotic treatments are often rendered ineffective due to the development of bacterial resistance, while Scutellaria barbata (SLB) has attracted widespread attention for its remarkable anti-inflammatory and antibacterial properties. However, its role in acne treatment has not been comprehensively studied. This study used high-performance liquid chromatography (HPLC) to analyze the bioactive components in a 70% ethanol extract of SLB. The antibacterial activity against C. acnes was systematically evaluated using well diffusion, minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and biofilm formation assays. Additionally, the effects of SLB on nitric oxide (NO) production and phagocytic activity were tested in RAW 264.7 cells. An acne skin model was established by treating HaCaT keratinocytes with heat-inactivated C. acnes. The results demonstrated that SLB significantly inhibited the growth of C. acnes and disrupted its biofilm formation. Moreover, SLB markedly reduced the secretion of inflammatory cytokines such as interleukin (IL)-6, IL-1β, IL-8, and tumor necrosis factor (TNF)-α in HaCaT keratinocytes stimulated by C. acnes. Moreover, SLB effectively alleviated skin barrier damage caused by C. acnes by suppressing the expression of matrix metalloproteinases (MMPs)-1, -3, -9, and -13. In conclusion, this study demonstrates that SLB possesses potent antibacterial, anti-inflammatory, and barrier-protective properties, making it a promising candidate for developing anti-acne products and exploring alternative antibiotic therapies. Full article
33 pages, 2636 KiB  
Article
From Traditional Medicine to the Laboratory: A Multidisciplinary Investigation on Agrimonia eupatoria L. Collected in Valle Imagna (BG, North of Italy)
by Fabrizia Milani, Chiara Muratore, Sara Biella, Martina Bottoni, Elio Rossi, Lorenzo Colombo, Paola Sira Colombo, Piero Bruschi, Alessio Papini, Paolo Landini, Claudia Giuliani, Fabrizio Araniti, Bhakti Prinsi and Gelsomina Fico
Plants 2025, 14(3), 340; https://doi.org/10.3390/plants14030340 - 23 Jan 2025
Viewed by 522
Abstract
A previous ethnobotanical investigation conducted in Valle Imagna (Northern Italy) highlighted the traditional use of Agrimonia eupatoria L. (Rosaceae) as a disinfectant and wound-healing agent. This use seemed to be linked to a local 18th century manuscript. This species was chosen for a [...] Read more.
A previous ethnobotanical investigation conducted in Valle Imagna (Northern Italy) highlighted the traditional use of Agrimonia eupatoria L. (Rosaceae) as a disinfectant and wound-healing agent. This use seemed to be linked to a local 18th century manuscript. This species was chosen for a multidisciplinary investigation to validate or refute its traditional use in the valley. Samples from fresh leaves were observed under Scanning Electron and Light Microscopy. The phenolic profiles of an epicuticular aqueous extract of the whole leaves and of infusions and decoctions of leaves and aerial parts were analyzed through Liquid Chromatography–Tandem Mass Spectrometry. The volatile organic compounds (VOCs) of fresh leaves were analyzed through Headspace Solid-Phase Microextraction coupled with Gas Chromatography–Mass Spectrometry. Growth inhibition and adhesion modulation were assessed on Escherichia coli, Staphylococcus aureus, and S. warneri by minimum inhibitory concentration and adhesion assays. Two trichome morphotypes were observed: a capitate with a one-celled rounded head and a capitate with a teo-celled cylindrical head. Both were responsible for producing terpenes, while the cylindrical capitates also produced polyphenols. Thirty-four phenolic compounds were characterized. Luteolin-7-O-glucoside, Catechin, and Epicatechin were common to all five extracts. The VOC profiles highlighted the dominance of (+)-α-Pinene. The infusions and the decoctions had a significant inhibitory activity on E. coli, and the extracts (specifically, the infusion of the leaves and both decoctions) also had a stimulating effect on the biofilm formation of S. warneri. These results already hold particular interest because of the strong connection they have to the traditional use of agrimony described in Valle Imagna. Full article
25 pages, 4657 KiB  
Article
Implant-Derived S. aureus Isolates Drive Strain-Specific Invasion Dynamics and Bioenergetic Alterations in Osteoblasts
by Lei Song, Lea-Sophie Schwinn, Juliane Barthel, Vanessa Ketter, Philipp Lechler, Uwe Linne, Ardawan J. Rastan, Sebastian Vogt, Steffen Ruchholtz, Jürgen R. J. Paletta and Madeline Günther
Antibiotics 2025, 14(2), 119; https://doi.org/10.3390/antibiotics14020119 - 23 Jan 2025
Viewed by 394
Abstract
Background: Implants are integral to modern orthopedic surgery. The outcomes are good, but infections remain a serious issue. Staphylococcus aureus (S. aureus), along with Staphylococcus epidermidis, are predominant pathogens responsible for implant-associated infections, as conventional antibiotic treatments often fail due [...] Read more.
Background: Implants are integral to modern orthopedic surgery. The outcomes are good, but infections remain a serious issue. Staphylococcus aureus (S. aureus), along with Staphylococcus epidermidis, are predominant pathogens responsible for implant-associated infections, as conventional antibiotic treatments often fail due to biofilm formation or the pathogens’ ability to invade cells and to persist intracellularly. Objectives: This study therefore focused on interactions of S. aureus isolates from infected implants with MG63 and SaOS2 osteoblasts by investigating the adhesion, invasion, and the impact on the bioenergetics of osteoblasts. Methods and Results: We found that the ability of S. aureus to adhere to osteoblasts depends on the isolate and was not associated with a single gene or expression pattern of characteristic adhesion proteins, and further, was not correlated with invasion. However, analysis of invasion capabilities identified better invasion conditions for S. aureus isolates with the SaOS2 osteoblastic cells. Interestingly, metabolic activity of osteoblasts remained unaffected by S. aureus infection, indicating cell survival. In contrast, respiration assays revealed an altered mitochondrial bioenergetic turnover in infected cells. While basal as well as maximal respiration in MG63 osteoblasts were not influenced statistically by S. aureus infections, we found increased non-mitochondrial respiration and enhanced glycolytic activity in the osteoblasts, which was again, more pronounced in the SaOS2 osteoblastic cells. Conclusions: Our findings highlight the complexity of S. aureus-host interactions, where both the pathogen and the host cell contribute to intracellular persistence and survival, representing a major factor for therapeutic failures. Full article
Show Figures

Figure 1

Figure 1
<p>Characterization of <span class="html-italic">S. aureus</span> isolates from implant associated infections. (<b>A</b>) Result of the API<sup>®</sup> Staph. test of isolates obtained from patients that underwent surgery for an implant-associated infection. (<b>B</b>) Color-changing strip of patient 42 is presented as an example. (<b>C</b>) Homologies of <span class="html-italic">S. aureus</span> isolates based on 16S rRNA sequencing, and (<b>D</b>) phylogenetic tree based on DNA likelihood with molecular clock. (<b>E</b>) Growth curve of the isolates in LB medium. (<b>F</b>) Biofilm formation of the isolates in LB and LB medium supplemented with NaCl and glucose. 0: blank; GLU: D-Glucose; FRU: D-Fructose; MNE: D-Mannose; MAL: D-Maltose; LAC: D-Lactose; TRE: D-Trehalose; MAN: D-Mannitol; XLT: Xylitol; MEL: D-Melibiose: NIT: Kaliumnitrat: PAL: ß-Naphthol-Phosphat; VP: Voges Proskauer; RAF: D-Raffinose; XYL: D-Xylose; SAC: D-Saccharose; MDG: Methyl-αD Glucopyranoside; NAG: N-acetyl-glucosamine; ADH: Arginine DiHydrolase; URE: UREase; +: Positive; −: Negative.</p>
Full article ">Figure 2
<p>Adhesion and invasion of <span class="html-italic">S. aureus</span> isolates on osteoblast like cell lines. (<b>A</b>) SaOS2 and MG63 were grown to confluence in 6-well plates and then incubated with <span class="html-italic">S. aureus</span> isolates from stationary growth phase. After 30 min, non-adherend bacteria were washed away with PBS. Then, osteoblasts were lysed and released bacteria were plated on LB-agar in order to determine colony-forming units. * <span class="html-italic">p</span> &lt; 0.05 compared to plastic surface (control). (<b>B</b>) Invasion assay of <span class="html-italic">S. aureus</span> isolates in SaOS2 and MG63 cell lines. After adhesion and washing, cells were incubated in DMEM supplemented with 10% FCS and 30 μg/mL gentamicin overnight in order to selectively eliminate remaining extracellular bacteria by quantifying colony-forming units. * <span class="html-italic">p</span> &lt; 0.05 compared to plastic surface (control). (<b>C</b>) Genes and proteins involved in the adhesion of <span class="html-italic">S. aureus</span> isolates as determined by PCR or mass spectrometry. (<b>D</b>) Genes for adhesion molecules found in <span class="html-italic">S. aureus</span> isolates were analyzed with respect to their expression level using qPCR; bbp encoding bone sialoprotein binding protein, cna collagen binding protein, eno laminin binding protein, fnbA and fnbB fibronectin binding proteins A and B, fib fibrinogen binding protein, clfA and clfB clumping factors A and B, ebpS elastin binding protein; psmA and psmB Phenol-soluble modulins A and B; eap extracellular adher-ence protein; sdrD SD-repeat containing protein D; fmtB methicillin resistance determinant FmtB protein; ebh extracellular matrix-binding protein.</p>
Full article ">Figure 3
<p>Cellular viability of osteoblasts after <span class="html-italic">S. aureus</span> infection. (<b>A</b>–<b>I</b>) Confluent layers of SaOS2 and MG63 osteoblasts were exposed to the different bacterial isolates (harvested during the stationary phase) for 30 min. Following incubation, non-adherent bacteria were removed by washing with PBS. Subsequently, DMEM supplemented with 10% FCS and 30 μg/mL gentamicin was applied to eliminate extracellular while preserving intracellular bacteria. After 1, 2, and 3 days metabolic activity was determined using MTT assay (+ <span class="html-italic">p</span> &lt; 0.05 indicating significant difference between SaOS2 and MG63 cells; * <span class="html-italic">p</span> &lt; 0.05 indicating a significant difference between single time points).</p>
Full article ">Figure 4
<p>Osteoblasts and <span class="html-italic">S. aureus</span> differ in their bioenergetic profiles. (<b>A</b>) Oxygen consumption rate (OCR) and (<b>B</b>) extracellular acidification rate (ECAR) of MG63 and SaOS2 cells. The OCR was measured under basal and stressed conditions using Seahorse technology with a Mito Stress Test by sequential injections of oligomycin (Oligo), FCCP, rotenone/antimycin A (Rot/AA) 24 h after seeding. (<b>C</b>–<b>F</b>) A calculation of mitochondrial activity was performed as illustrated. Maximal respiration resulted from the OCR after the injection of FCCP minus the OCR after injection of oligomycin. Non-mitochondrial oxygen consumption rates were obtained by the OCR levels after Rot/AA injection. (<b>G</b>,<b>H</b>) Respiratory activity of <span class="html-italic">S. aureus</span> ATCC<sup>®</sup> 29213<sup>TM</sup> in osteoblast-free conditions after 24 h and (<b>I</b>–<b>L</b>) metabolic parameters calculated in accordance with (<b>A</b>), respectively. genta: gentamicin.</p>
Full article ">Figure 5
<p>Mitochondrial respiration is altered in MG63 osteoblasts infected with <span class="html-italic">S. aureus</span> isolates. (<b>A</b>,<b>B</b>,<b>G</b>,<b>H</b>) MG63 cells were infected with isolates from patients 2, 4, 6, 9, 28, 36, 42, 45, and <span class="html-italic">S. aureus</span> ATCC<sup>®</sup> 29213<sup>TM</sup>, followed by gentamycin supplementation in order to remove extracellular bacteria. After one day, respiration was analyzed by a Seahorse Mito Stress Test, followed by the quantification of the basal respiration (<b>C</b>,<b>I</b>), non-mitochondrial oxygen consumption (<b>D</b>,<b>J</b>), maximal respiration (<b>E</b>,<b>K</b>), and basal ECAR (<b>F</b>,<b>L</b>) from the obtained respiratory profiles. All results are representative of at least three independent experiments. Data are demonstrated as mean ± SD of at least six replicate samples. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to uninfected control, ANOVA, Bonferroni’s test.</p>
Full article ">Figure 6
<p>Mitochondrial respiration is altered in SaOS2 osteoblasts infected with <span class="html-italic">S. aureus</span> isolates. (<b>A</b>,<b>B</b>,<b>G</b>,<b>H</b>) SaOS2 cells were infected with isolates from patients 2, 4, 6, 9, 28, 36, 42, 45 and <span class="html-italic">S. aureus</span> ATCC<sup>®</sup> 29213<sup>TM</sup>, followed by gentamycin supplementation in order to remove extracellular bacteria. After day one, respiration was analyzed by a Seahorse Mito Stress Test, followed by the quantification of the basal respiration (<b>C</b>,<b>I</b>), non-mitochondrial oxygen consumption (<b>D</b>,<b>J</b>), maximal respiration (<b>E</b>,<b>K</b>), and basal ECAR (<b>F</b>,<b>L</b>) from the respective respiratory profiles. All results are representative of at least three independent experiments. Data are demonstrated as mean ± SD of at least six replicate samples. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to uninfected control, ANOVA, Bonferroni’s test.</p>
Full article ">
15 pages, 4054 KiB  
Article
Antibiofilm Activity of Protamine Against the Vaginal Candidiasis Isolates of Candida albicans, Candida tropicalis and Candida krusei
by Sivakumar Jeyarajan, Indira Kandasamy, Raja Veerapandian, Jayasudha Jayachandran, Shona Chandrashekar, Kalimuthusamy Natarajaseenivasan, Prahalathan Chidambaram and Anbarasu Kumarasamy
Appl. Biosci. 2025, 4(1), 5; https://doi.org/10.3390/applbiosci4010005 - 23 Jan 2025
Viewed by 359
Abstract
Candida species, normally part of the healthy human flora, can cause severe opportunistic infections when their population increases. This risk is even greater in immunocompromised individuals. Women using intrauterine contraceptive devices (IUDs) are at higher risk for IUD-associated vulvovaginal candidiasis (VVC) because the [...] Read more.
Candida species, normally part of the healthy human flora, can cause severe opportunistic infections when their population increases. This risk is even greater in immunocompromised individuals. Women using intrauterine contraceptive devices (IUDs) are at higher risk for IUD-associated vulvovaginal candidiasis (VVC) because the device provides a surface for biofilm formation. This biofilm formation allows the normal flora to become opportunistic pathogens, leading to symptoms of VVC such as hemorrhage, pelvic pain, inflammation, itching and discharge. VVC is often linked to IUD use, requiring the prompt removal of these devices for effective treatment. This study evaluated the activity of the arginine-rich peptide “protamine” against Candida albicans, Candida tropicalis and Candida krusei isolated from IUD users who had signs of VVC. The antimicrobial activity was measured using the agar disk diffusion and microbroth dilution methods to determine the minimum inhibitory concentration (MIC). The MIC values of protamine against C. albicans, C. tropicalis and C. krusei are 32 μg mL−1, 64 μg mL−1 and 256 μg mL−1, respectively. The determined MIC of protamine was used for a biofilm inhibition assay by crystal violet staining. Protamine inhibited the biofilm formation of the VVC isolates, and its mechanisms were studied through scanning electron microscopy (SEM) and a reactive oxygen species (ROS) assay. The disruption of cell membranes and the induction of oxidative stress appear to be key mechanisms underlying its anti-candidal effects. The results from an in vitro assay support the potential use of protamine as an antibiofilm agent to coat IUDs in the future for protective purposes. Full article
Show Figures

Figure 1

Figure 1
<p>Susceptibility of (<b>a</b>) <span class="html-italic">C. albicans</span> (MTCC 227), VVC <span class="html-italic">Candida</span> spp. isolates, (<b>b</b>) <span class="html-italic">C. tropicalis</span> (CA4) and (<b>c</b>) <span class="html-italic">C. krusei</span> (CA54) to protamine measured by an agar diffusion assay. The numbers marked on the plate images refer to the concentration of protamine loaded onto the disks (1–10, 2–20, 3–30, 4–40 and 5–50 μg). The inset shows the image of a disk loaded with PBS as a control. The bar graphs (<b>d</b>–<b>f</b>) below each image show the diameter of the zone of inhibition (ZOI) plotted for each concentration for each species. The control shows the diameter of the disk alone (0.6 cm). For <span class="html-italic">C. albicans</span> and <span class="html-italic">C. krusei</span>, a concentration gradient increase in the ZOI was observed. For C. <span class="html-italic">krusei</span>, no ZOI was observed for the measured concentrations of 10 to 50 µg.</p>
Full article ">Figure 2
<p><span class="html-italic">Candida</span>-cidal activity of protamine against <span class="html-italic">Candida</span> spp. (<b>a</b>–<b>c</b>). A microbroth dilution assay was performed from 0 to 512 µg mL<sup>−1</sup> to determine the MIC. Cell densities for <span class="html-italic">Candida</span> spp. measured at 595 nm without protamine are used as controls with 100% growth. The MIC of protamine is 32 µg mL<sup>−1</sup> against <span class="html-italic">C. albicans</span>, 64 µg mL<sup>−1</sup> against <span class="html-italic">C. tropicalis</span> and 256 µg mL<sup>−1</sup> against <span class="html-italic">C. krusei</span>.</p>
Full article ">Figure 3
<p>Light microscopic image of crystal violet-stained control cells: (<b>a</b>) <span class="html-italic">C. albicans</span>, (<b>b</b>) <span class="html-italic">C. tropicalis</span> (CA4) and (<b>c</b>) <span class="html-italic">C. Krusei</span> (CA54) at 40× magnification. The cells (<b>d</b>–<b>f</b>) were treated with 32, 64 and 256 µg mL<sup>−1</sup> of protamine, respectively, for 24 h and stained with crystal violet.</p>
Full article ">Figure 4
<p>Scanning electron microscopic (SEM) image of <span class="html-italic">C. albicans</span> (MTCC 227): (<b>a</b>) the control and (<b>b</b>) protamine-treated cells. <span class="html-italic">C. albicans</span> was treated with protamine at 32 µg mL<sup>−1</sup> for 6 h. A reduction in the number of cells is noted as well as empty spaces between the cells and the disruption of the cell wall, demonstrating the lytic activity of protamine. The protamine-treated cells show plasmolysis and a ruptured surface, with internally collapsed morphology due to cytoplasm leakage and a shrunk appearance with grooves compared to the control cells.</p>
Full article ">Figure 5
<p>Effect of protamine on ROS production in <span class="html-italic">Candida</span> spp. Protamine at its MIC was added to VVC <span class="html-italic">Candida</span> spp. isolates, and ROS generation was studied using DCFH-DA. The nonfluorescent dye DCFH-DA at a concentration of 10 μM was pre-incubated with <span class="html-italic">Candida</span> spp. and allowed to grow for 24 h at 37 °C with and without protamine. After 24 h, the culture dish was washed with PBS (pH 7.4) and observed under a fluorescent microscope. The images were captured at an excitation wavelength of 488 nm and an emission wavelength of 525 nm for the DCFH-DA. The generation of ROS will result in DCFH-DA fluorescence. Although the control cells did not exhibit any fluorescence, the cells treated with protamine at their MIC displayed green fluorescence. Hoechst dye was used to stain the nucleus and appears blue. For protamine-treated cells, there is less blue fluorescence (both in the number of cells and intensity), showing signs of a damaged nucleus. The top two rows of panels show <span class="html-italic">C. albicans</span> without (<b>a</b>–<b>d</b>) and with (<b>e</b>–<b>h</b>) protamine. The two middle rows of panels represent <span class="html-italic">C. tropicalis</span> treated with (<b>m</b>–<b>p</b>) and without (<b>i</b>–<b>l</b>) protamine. C. <span class="html-italic">krusei</span> with (<b>u</b>–<b>x</b>) and without (<b>q</b>–<b>t</b>) protamine is shown in the bottom two rows of panels.</p>
Full article ">
19 pages, 2947 KiB  
Article
Redox-Modulating Capacity and Effect of Ethyl Acetate Roots and Aerial Parts Extracts from Geum urbanum L. on the Phenotype Inhibition of the Pseudomonas aeruginosa Las/RhI Quorum Sensing System
by Lyudmila Dimitrova, Milka Mileva, Almira Georgieva, Elina Tzvetanova, Milena Popova, Vassya Bankova and Hristo Najdenski
Plants 2025, 14(2), 213; https://doi.org/10.3390/plants14020213 - 14 Jan 2025
Viewed by 529
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes nosocomial infections of the urinary tract, upper respiratory tract, gastrointestinal tract, central nervous system, etc. It is possible to develop bacteremia and sepsis in immunocompromised patients. A major problem in treatment is the development of [...] Read more.
Pseudomonas aeruginosa is an opportunistic pathogen that causes nosocomial infections of the urinary tract, upper respiratory tract, gastrointestinal tract, central nervous system, etc. It is possible to develop bacteremia and sepsis in immunocompromised patients. A major problem in treatment is the development of antibiotic resistance. Therefore, new preparations of natural origin are sought, such as plant extracts, which are phytocomplexes and to which it is practically impossible to develop resistance. Geum urbanum L. (Rosacea) is a perennial herb known for many biological properties. This study aimed to investigate the redox-modulating capacity and effect of ethyl acetate (EtOAc) extracts from roots (EtOAcR) and aerial parts (EtOAcAP) of the Bulgarian plant on the phenotype inhibition of the P. aeruginosa Las/RhI quorum sensing (QS) system, which primarily determines drug resistance in pathogenic bacteria, including biofilm formation, motility, and pigment production. We performed QS assays to account for the effects of the two EtOAc extracts. At sub-minimal inhibitory concentrations (sub-MICs) ranging from 1.56 to 6.25 mg/mL, the biofilm formation was inhibited 85% and 84% by EtOAcR and 62% and 39% by EtOAcAP extracts, respectively. At the same sub-MICs, the pyocyanin synthesis was inhibited by 17–27% after treatment with EtOAcAP and 26–30% with EtOAcR extracts. The motility was fully inhibited at 3.12 mg/mL and 6.25 mg/mL (sub-MICs). We investigated the inhibitory potential of lasI, lasR, rhiI, and rhiR gene expression in biofilm and pyocyanin probes with the PCR method. Interestingly, the genes were inhibited by two extracts at 3.12 mg/mL and 6.25 mg/mL. Antiradical studies, assessed by DPPH, CUPRAC, and ABTS radical scavenging methods and superoxide anion inhibition showed that EtOAcAP extract has effective antioxidant capacity. These results could help in the development of new phytocomplexes that could be applied as biocontrol agents to inhibit the phenotype of the P. aeruginosa QS system and other antibiotic-resistant pathogens. Full article
(This article belongs to the Section Plant Protection and Biotic Interactions)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram showing gene regulation for the Las, Rhl, and PQS systems in <span class="html-italic">P. aeruginosa</span>. Legend: ‘’+’’—activation, ‘’-‘’—inhibition, Vfr—regulator of the virulence factor, GacA—global activator, LasR (<span class="html-italic">lasR</span> gene)—transcription activator, RhlR (<span class="html-italic">rhIR</span> gene)—transcription activator, LasI (<span class="html-italic">lasI</span> gene)—synthase of autoinducer 3-oxo-C<sub>12</sub>-HSL, RhII (<span class="html-italic">rhII</span> gene)—synthase of autoinducer C<sub>4</sub>-HSL, 3-oxo-C12-HSL—N-3-oxododecanoyl-homoserine lactone, C<sub>4</sub>-HSL—N-butyryl-homoserine lactone, PQS—<span class="html-italic">Pseudomonas</span> quinolone system (autoinducer is 2-heptyl-3-hydroxy-4-Quinolone), and pqsABCDE—operon that promotes PQS production.</p>
Full article ">Figure 2
<p>Biofilm formation of <span class="html-italic">P. aeruginosa</span> untreated and treated with EtOAcAP and EtOAcR from <span class="html-italic">G. urbanum</span>.</p>
Full article ">Figure 3
<p>Pyocyanin production quantity of <span class="html-italic">P. aeruginosa</span> untreated and treated with EtOAcAP and EtOAcR from <span class="html-italic">G. urbanum</span>.</p>
Full article ">Figure 4
<p>Swarming motility capacity of P. aeruginosa untreated and treated with EtOAcAP and EtOAcR from <span class="html-italic">G. urbanum</span>. Legend: (<b>A</b>) untreated control, (<b>B</b>) treated bacterial culture with 6.25 mg/mL of EtOAcAP extract, (<b>C</b>) treated bacterial culture with 3.12 mg/mL of EtOAcAP extract, (<b>D</b>) treated bacterial culture with 6.25 mg/mL of EtOAcR extract, and (<b>E</b>) treated bacterial culture with 3.12 mg/mL of EtOAcR extract.</p>
Full article ">Figure 5
<p>Las/RhI regulatory system gene expression by PCR from biofilm-forming and pyocyanin production probes. (<b>A</b>) <span class="html-italic">rhI</span>I and <span class="html-italic">rhI</span>R genes (M—marker; Co(−)—negative control; Co(+)—positive control. (1) EtOAcAP 6.25 mg/mL, (2) EtOAcAP 3.12 mg/mL, (3) EtOAcR 6.25 mg/mL, and (4) EtOAc 3.12 mg/mL). (<b>B</b>) <span class="html-italic">las</span>I and <span class="html-italic">las</span>R genes (M—marker; Co(−)—negative control; Co(+)—positive control. (1) EtOAcAP 6.25 mg/mL, (2) EtOAcAP 3.12 mg/mL, (3) EtOAcR 6.25 mg/mL, and (4) EtOAc 3.12 mg/mL).</p>
Full article ">Figure 5 Cont.
<p>Las/RhI regulatory system gene expression by PCR from biofilm-forming and pyocyanin production probes. (<b>A</b>) <span class="html-italic">rhI</span>I and <span class="html-italic">rhI</span>R genes (M—marker; Co(−)—negative control; Co(+)—positive control. (1) EtOAcAP 6.25 mg/mL, (2) EtOAcAP 3.12 mg/mL, (3) EtOAcR 6.25 mg/mL, and (4) EtOAc 3.12 mg/mL). (<b>B</b>) <span class="html-italic">las</span>I and <span class="html-italic">las</span>R genes (M—marker; Co(−)—negative control; Co(+)—positive control. (1) EtOAcAP 6.25 mg/mL, (2) EtOAcAP 3.12 mg/mL, (3) EtOAcR 6.25 mg/mL, and (4) EtOAc 3.12 mg/mL).</p>
Full article ">
24 pages, 20737 KiB  
Article
Antimicrobial Efficacy of Trifluoro-Anilines Against Vibrio Species
by Ezhaveni Sathiyamoorthi, Bharath Reddy Boya, Jin-Hyung Lee and Jintae Lee
Int. J. Mol. Sci. 2025, 26(2), 623; https://doi.org/10.3390/ijms26020623 - 13 Jan 2025
Viewed by 284
Abstract
Vibrios are naturally present in marine ecosystems and are commonly allied with live seafood. Vibrio species frequently cause foodborne infections, with Vibrio parahaemolyticus recently becoming a significant contributor to foodborne illness outbreaks. In response, aniline and 68 of its aniline derivatives were studied [...] Read more.
Vibrios are naturally present in marine ecosystems and are commonly allied with live seafood. Vibrio species frequently cause foodborne infections, with Vibrio parahaemolyticus recently becoming a significant contributor to foodborne illness outbreaks. In response, aniline and 68 of its aniline derivatives were studied due to their antibacterial effects targeting V. parahaemolyticus and Vibrio harveyi. Among these, 4-amino-3-chloro-5-nitrobenzotrifluoride (ACNBF) and 2-iodo-4-trifluoromethylaniline (ITFMA) demonstrated both antibacterial and antibiofilm properties. The minimum inhibitory concentrations (MIC) for ACNBF and ITFMA were 100 µg/mL and 50 µg/mL, respectively, against planktonic cells. The active compounds effectively suppressed biofilm formation in a manner dependent on the dosage. Additionally, these trifluoro-anilines significantly reduced virulence factors such as motility, protease activity, hemolysis, and indole production. Both trifluoro-anilines caused noticeable destruction to the membrane of bacterial cells and, at 100 µg/mL, exhibited bactericidal activity against V. parahaemolyticus within 30 min. Toxicity assays using the Caenorhabditis elegans and seed germination models showed that the compounds displayed mild toxicity. As a result, ACNBF and ITFMA inhibited the growth of both planktonic cells and biofilm formation. Furthermore, these active compounds effectively prevented the formation of biofilm on the surfaces of shrimp and squid models, highlighting their potential use in controlling seafood contamination. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of 68 aniline derivatives on inhibiting biofilm formation by <span class="html-italic">V. parahaemolyticus</span> (<b>A</b>) and <span class="html-italic">V. harveyi</span> (<b>B</b>). Numbers 1–68 refer to aniline and its derivatives, as outlined in <a href="#ijms-26-00623-t001" class="html-table">Table 1</a>. * <span class="html-italic">p</span> &lt; 0.05 vs. non-treated controls.</p>
Full article ">Figure 2
<p>The activity of inhibiting biofilm formation in <span class="html-italic">V. parahaemolyticus</span> by aniline and its two hit compounds 4-amino-3-chloro-5-nitrobenzotrifluoride (ACNBF) and 2-iodo-4-(trifluoromethylaniline) (ITFMA) (<b>A</b>). The cell growth pattern of <span class="html-italic">V. parahaemolyticus</span> exposed to ACNBF and ITFMA aniline derivatives (<b>B</b>). Protease activity evaluation (<b>C</b>). The indole was analyzed at pH 7 (<b>D</b>), hemolysin assay (<b>E</b>). The asterisk (*) indicates statistical significance cells with a <span class="html-italic">p</span>-value of less than 0.05.</p>
Full article ">Figure 3
<p>Impact of active compounds on the bacterial cell motility of <span class="html-italic">V. parahaemolyticus</span>. Swim movement (<b>A</b>) and swarm movement (<b>B</b>). The swimming (<b>C</b>) and swarming (<b>D</b>) diameter measurement of two active aniline derivatives. * <span class="html-italic">p</span> &lt; 0.05 vs. non-treated controls.</p>
Full article ">Figure 4
<p>Microscopic evaluation of <span class="html-italic">V. parahaemolyticus</span> biofilms by using aniline derivatives (<b>A</b>). SEM imaging revealed biofilm cells treated with aniline, ACNBF, and ITFMA (100 μg/mL) (<b>B</b>). The colors of yellow and white bars indicate sizes of 6 and 1.5 µm, respectively.</p>
Full article ">Figure 5
<p>Time-killing assay with active aniline derivatives against <span class="html-italic">V. parahaemolyticus</span> (<b>A</b>). Aniline derivatives demonstrated antibacterial efficacy in a cooked shrimp model (<b>B</b>). SEM images demonstrate that aniline derivatives, at 100 µg/mL concentrations of ACNBF and ITFMA, eradicate bacteria on squid surfaces (<b>C</b>). The measure bars displayed in red and blue scale bars represent of 1.5 µm and 6 µm, respectively.</p>
Full article ">Figure 6
<p>Production of ROS in V. parahaemolyticus. The blue bars represent the effects of the compounds (Aniline (<b>A</b>), ACNBF (<b>B</b>), and ITFMA (<b>C</b>)), while the gray bars indicate the effect of H<sub>2</sub>O<sub>2</sub> at comparable concentrations (used as a reference for oxidative stress). * <span class="html-italic">p</span> &lt; 0.05 vs. non-treated controls.</p>
Full article ">Figure 7
<p>Effect of aniline and its derivatives on <span class="html-italic">Brassica rapa</span> growth over 7 days at concentrations of 10–400 μg/mL (<b>A</b>), and toxicity assay evaluating <span class="html-italic">C. elegans</span> survival following treatment with aniline and its derivatives at 10–100 μg/mL for 7 days (<b>B</b>).</p>
Full article ">
22 pages, 7293 KiB  
Article
Cinnamic Acid Compounds (p-Coumaric, Ferulic, and p-Methoxycinnamic Acid) as Effective Antibacterial Agents Against Colistin-Resistant Acinetobacter baumannii
by Alaaddin Korkut, Serap Özkaya Gül, Esra Aydemir, Hakan Er and Elif Odabaş Köse
Antibiotics 2025, 14(1), 71; https://doi.org/10.3390/antibiotics14010071 - 11 Jan 2025
Viewed by 721
Abstract
Colistin-resistant Acinetobacter baumannii (COLR-Ab) is an opportunistic pathogen commonly associated with nosocomial infections, and it is difficult to treat with current antibiotics. Therefore, new antimicrobial agents need to be developed for treatment. Based on this information, we investigated the antimicrobial, antibiofilm, and combination [...] Read more.
Colistin-resistant Acinetobacter baumannii (COLR-Ab) is an opportunistic pathogen commonly associated with nosocomial infections, and it is difficult to treat with current antibiotics. Therefore, new antimicrobial agents need to be developed for treatment. Based on this information, we investigated the antimicrobial, antibiofilm, and combination activities of p-coumaric acid (p-CA), ferulic acid (FA), and p-methoxycinnamic acid (p-MCA) against five COLR-Ab isolates. p-CA, FA, and p-MCA exhibited antimicrobial activity against COLR-Ab isolates, with minimum inhibitory concentration (MIC) values in the range of 256–128 µg/mL, 1024–512 µg/mL, and 512–128 µg/mL, respectively. The combination effects of the compounds with colistin (COL) were evaluated using a checkerboard synergy test. The combinations exhibited a synergistic effect and caused a 128- to 16-fold decrease in COL MIC values. In addition, the biofilm production capacities of the COLR-Ab isolates and the antibiofilm activities of the compounds were determined using the microtitre plate-based crystal violet (CV) technique. The compounds showed effective antibiofilm activity against strong and moderate biofilm-producing isolates, inhibiting biofilm formation by 77.5% and 19.7%. Spectrometric measurements were used to examine the effect of compounds on membrane permeability; 1.9-, 1.66-, and 1.34-fold increases in absorbance values were observed at MIC concentrations of p-CA, FA, and p-MCA, respectively. Furthermore, morphological changes caused by the compounds in the isolate were observed using scanning electron microscopy (SEM) micrographs. According to the WST assay, the compounds did not show any statistically significant cytotoxic effect on the cells (p > 0.05). These findings indicate that p-CA, FA, and p-MCA may be potential new alternative candidates against resistant A. baumannii. Full article
Show Figures

Figure 1

Figure 1
<p>Biofilm formation ability of COLR-Ab isolates. *** <span class="html-italic">p</span> ≤ 0.001 and **** <span class="html-italic">p</span> ≤ 0.0001 were considered to indicate statistically significant differences compared to the control group.</p>
Full article ">Figure 2
<p>Antibiofilm activities of COL (<b>A</b>), <span class="html-italic">p</span>-CA (<b>B</b>), FA (<b>C</b>), and <span class="html-italic">p</span>-MCA (<b>D</b>) against COLR-Ab1 isolate. * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001 were considered to indicate statistically significant differences compared to the growth control group. ns: not significant (<span class="html-italic">p</span> &gt; 0.05); C, control; Comb, combination.</p>
Full article ">Figure 3
<p>Antibiofilm activities of COL (<b>A</b>), <span class="html-italic">p</span>-CA (<b>B</b>), FA (<b>C</b>), and <span class="html-italic">p</span>-MCA (<b>D</b>) against COLR-Ab3 isolate. * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001 were considered to indicate statistically significant differences compared to the growth control group. ns: not significant (<span class="html-italic">p</span> &gt; 0.05); C, control; Comb, combination.</p>
Full article ">Figure 4
<p>Antibiofilm activities of COL (<b>A</b>), <span class="html-italic">p</span>-CA (<b>B</b>), FA (<b>C</b>), and <span class="html-italic">p</span>-MCA (<b>D</b>) against COLR-Ab4 isolate. **** <span class="html-italic">p</span> ≤ 0.0001 were considered to indicate statistically significant differences compared to the growth control group. ns: not significant (<span class="html-italic">p</span> &gt; 0.05); C, control; Comb, combination.</p>
Full article ">Figure 5
<p>Antibiofilm activities of COL (<b>A</b>), <span class="html-italic">p</span>-CA (<b>B</b>), FA (<b>C</b>) and <span class="html-italic">p</span>-MCA (<b>D</b>) against COLR-Ab5 isolate. * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001 were considered to indicate statistically significant differences compared to the growth control group. ns: not significant (<span class="html-italic">p</span> &gt; 0.05); C, control; Comb, combination.</p>
Full article ">Figure 6
<p>Absorbance values obtained by treating concentrations of <span class="html-italic">p</span>-CA, FA, and <span class="html-italic">p</span>-MCA at MIC, MIC/2, and MIC/4 values against the COLR-Ab4 isolate. ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001 were considered to indicate statistically significant differences compared to the control group. ns: not significant (<span class="html-italic">p</span> &gt; 0.05); C, control.</p>
Full article ">Figure 7
<p>Scanning electron micrographs of the COLR-Ab4 isolate in the presence of MIC concentrations of <span class="html-italic">p</span>-CA, FA, and <span class="html-italic">p</span>-MCA: (<b>A</b>) untreated; (<b>B</b>) treated with <span class="html-italic">p</span>-CA; (<b>C</b>) treated with FA; (<b>D</b>) treated with <span class="html-italic">p</span>-MCA. White arrows indicate damaged bacterial cells. Scale bars, 1 µm in (<b>A</b>,<b>C</b>) and 2 µm in (<b>B</b>,<b>D</b>).</p>
Full article ">Figure 8
<p>Effect of (<b>A</b>) etoposide, (<b>B</b>) <span class="html-italic">p</span>-CA, (<b>C</b>) FA, and (<b>D</b>) <span class="html-italic">p</span>-MCA on cell viability in 293T cells after 24 h of incubation. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 were considered to indicate statistically significant differences compared to the control group.</p>
Full article ">Figure 9
<p>Effect of (<b>A</b>) etoposide, (<b>B</b>) <span class="html-italic">p</span>-CA, (<b>C</b>) FA, and (<b>D</b>) <span class="html-italic">p</span>-MCA on cell viability in HUVEC cells after 24 h of incubation. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 were considered to indicate statistically significant differences compared to the control group.</p>
Full article ">Figure 10
<p>Effect of (<b>A</b>) etoposide, (<b>B</b>) <span class="html-italic">p</span>-CA, (<b>C</b>) FA, and (<b>D</b>) <span class="html-italic">p</span>-MCA on cell viability in Vero cells after 24 h of incubation. * <span class="html-italic">p</span> &lt; 0.05 was considered to indicate statistically significant differences compared to the control group.</p>
Full article ">
25 pages, 2475 KiB  
Article
Green Tea Extract (Theaceae; Camellia sinensis L.): A Promising Antimicrobial, Anti-Quorum Sensing and Antibiofilm Candidate Against Multidrug-Resistant Campylobacter Species
by Mona S. Emara, Ahmed M. Ammar, Ashraf M.O. Abdelwahab, Attia A. Elgdawy, Adel Abdelkhalek, Elena Pet, Gabi Dumitrescu, Mirela Ahmadi and Norhan K. Abd El-Aziz
Antibiotics 2025, 14(1), 61; https://doi.org/10.3390/antibiotics14010061 - 9 Jan 2025
Viewed by 734
Abstract
Background: Thermophilic Campylobacter species are among the main culprits behind bacterial gastroenteritis globally and have grown progressively resistant to clinically important antimicrobials. Many studies have been carried out to explore innovative and alternative strategies to control antibiotic-resistant campylobacters in animal reservoirs and human [...] Read more.
Background: Thermophilic Campylobacter species are among the main culprits behind bacterial gastroenteritis globally and have grown progressively resistant to clinically important antimicrobials. Many studies have been carried out to explore innovative and alternative strategies to control antibiotic-resistant campylobacters in animal reservoirs and human hosts; however, limited studies have been performed to develop efficient control schemes against Campylobacter biofilms. Methods: This study investigated the antimicrobial and antibiofilm activities of some herbal extracts against multidrug-resistant (MDR) Campylobacter species recovered from different sources using phenotypic and molecular techniques. Results: The overall Campylobacter species prevalence was 21.5%, representing 15.25% and 6.25% for C. jejuni and C. coli, respectively. Regarding C. jejuni, the highest resistance rate was observed for amoxicillin–clavulanic acid and colistin (85.25% each), followed by cefotaxime (83.61%) and tetracycline (81.97%), whereas C. coli isolates showed absolute resistance to cefotaxime followed by erythromycin (92%) and colistin (88%). Remarkably, all Campylobacter isolates were MDR with elevated multiple antimicrobial resistance (MAR) indices (0.54–1). The antimicrobial potentials of green tea (Camellia sinensis), rosemary (Rosmarinus officinalis) and ginger (Zingiber officinale) extracts against MDR Campylobacter isolates were assessed by the disk diffusion assay and broth microdilution technique. Green tea extract showed a marked inhibitory effect against tested isolates, exhibiting growth inhibition zone diameters of 8 to 38 mm and a minimum inhibitory concentration (MIC) range of 1.56–3.12 mg/mL, unlike the rosemary and ginger extracts. Our findings reveal a respectable antibiofilm activity (>50% biofilm formation inhibition) of green tea against the preformed biofilms of Campylobacter isolates. Furthermore, real-time quantitative polymerase chain reaction (RT-qPCR) results showed a significant decrease (p < 0.05) in the expression levels of biofilm biosynthesis gene and its regulator (FlaA and LuxS, respectively) in Campylobacter isolates treated with the green tea extract in comparison with untreated ones. Conclusion: This is the first in vitro approach that has documented the inhibitory activity of green tea extract against MDR-biofilm-producing Campylobacter species isolated from different sources. Further in vivo studies in animals’ models should be performed to provide evidence of concept for the implementation of this alternative candidate for the mitigation of MDR Campylobacter infections in the future. Full article
(This article belongs to the Special Issue Antimicrobial and Antibiofilm Activity by Natural Compounds)
Show Figures

Figure 1

Figure 1
<p>The heatmap illustrates the antimicrobial resistance of <span class="html-italic">Campylobacter</span> species recovered from different sources. Dark colors refer to the antimicrobials displaying high resistance levels and light colors refers to the antimicrobials displaying low resistance. Color bar on the right side indicates color intensity. AMA, antimicrobial agent; P, penicillin; SAM, ampicillin–sulbactam; AMC, amoxicillin–clavulanic acid; E, erythromycin; TL, tylosin; CT, colistin; C, chloramphenicol; CTX, cefotaxime; TE, tetracycline; CN, gentamycin; CIP, ciprofloxacin; NOR, norfloxacin; SXT, sulfamethoxazole–trimethoprim. *** indicate significant differences at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>LC-ESI-MS/MS multiple reaction monitoring (MRM) chromatogram of the standard sample (<b>a</b>) and green tea extract sample (<b>b</b>). Different colours indicate extract constituents, and each analyte has two fragments generated at the same extension time. The scientific notation (i.e., 1.0e+6 is equal to 1 × 10<sup>6</sup>) could be converted into decimal notation using the following website: <a href="https://converthere.com/numbers/2.3e+7-written-out" target="_blank">https://converthere.com/numbers/2.3e+7-written-out</a> (accessed on 2 December 2024).</p>
Full article ">Figure 3
<p>Catechins (<b>a</b>) and epigallocatechin gallate [M-H2O-H]- (EGCG) (<b>b</b>) identified in the green tea extract by LC-ESI-MS/MS in negative mode. The scientific notation (i.e., 1.0e6 is equal to 1 × 10<sup>6</sup>) could be converted into decimal notation using the following website: <a href="https://converthere.com/numbers/2.3e+7-written-out" target="_blank">https://converthere.com/numbers/2.3e+7-written-out</a> (accessed on 2 December 2024).</p>
Full article ">Figure 4
<p>Epicatechin–gallate (ECG) (<b>a</b>) and epigallocatechine (EGC) (<b>b</b>) identified in the green tea extract by LC-ESI-MS/MS in positive mode. The scientific notation (i.e., 1e4 is equal to 1 × 10<sup>4</sup>) could be converted into decimal notation using the following website: <a href="https://converthere.com/numbers/2.3e+7-written-out" target="_blank">https://converthere.com/numbers/2.3e+7-written-out</a> (accessed on 2 December 2024).</p>
Full article ">Figure 5
<p>Fold changes in the expression levels of examined biofilm genes after treatment of <span class="html-italic">Campylobacter</span> isolates with sub-inhibitory concentrations of green tea extract. a–c Values of the same row with different superscripts are significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
17 pages, 3157 KiB  
Article
Relationships Between Candida auris and the Rest of the Candida World—Analysis of Dual-Species Biofilms and Infections
by Monika Janeczko and Tomasz Skrzypek
Pathogens 2025, 14(1), 40; https://doi.org/10.3390/pathogens14010040 - 8 Jan 2025
Viewed by 532
Abstract
In this study, we investigated the interactions between Candida auris and C. albicans, C. tropicalis, C. glabrata, and C. krusei in mixed infections. Initially, these interactions were studied qualitatively and quantitatively in dual-species biofilms formed in vitro. The MTT assays, [...] Read more.
In this study, we investigated the interactions between Candida auris and C. albicans, C. tropicalis, C. glabrata, and C. krusei in mixed infections. Initially, these interactions were studied qualitatively and quantitatively in dual-species biofilms formed in vitro. The MTT assays, determination of the total CFU/mL, and SEM analysis showed that C. auris interacted differentially with the other Candida spp. during the dual-species biofilm formation. Depending on the stage of the biofilm development, C. auris was found to be a particularly dominant species during its interaction with the C. krusei biofilms but significantly submissive in the C. auris-C. albicans biofilms. These studies were then extended to in vivo host models of experimental candidiasis. G. mellonella larvae were inoculated with monotypic and heterotypic suspensions of Candida. The survival rates and quantification of fungal cells in the hemolymph showed that the highest mortality was exhibited by larvae in the C. auris-C. albicans co-infection (100% mortality after 36 h). The CFU/mL values of C. auris from the larval hemolymph were lower in the interactive groups compared to the mono-species group. As a newly emerging species, C. auris persists in environments in the presence of other Candida species and is involved in both competitive and noncompetitive interactions with other Candida species during biofilm formation and development of experimental candidiasis. Full article
Show Figures

Figure 1

Figure 1
<p>Biofilm formation by <span class="html-italic">Candida</span> mono-species on microtitre plates. Biofilm viability was quantified by the MTT assay. At each time point of biofilm maturation, the optical density of <span class="html-italic">Candida</span> mono-species biofilms was compared with the optical density of <span class="html-italic">C. auris</span> (black columns, ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001) by a Student’s <span class="html-italic">t</span>-test. Data are the means ± SD of experiments performed in three duplicates and repeated two times.</p>
Full article ">Figure 2
<p>Biofilm formation of <span class="html-italic">Candida</span> dual-species on microtiter plates: (<b>a</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. albicans</span>, (<b>b</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. glabrata</span>, (<b>c</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. tropicalis</span>, and (<b>d</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. krusei</span>. Biofilm viability was quantified by the MTT assay. At each time point of biofilm maturation, the optical density of mixed biofilms was compared with the optical density of <span class="html-italic">C. auris</span> or non-auris <span class="html-italic">Candida</span> species (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001) by Student’s <span class="html-italic">t</span>-test. Data are the means ± SD of experiments performed in three duplicates and repeated three times.</p>
Full article ">Figure 3
<p>Representative scanning electron microscopy images of dual-species <span class="html-italic">Candida</span> biofilms compared with the mono-species <span class="html-italic">C. auris</span> biofilm after 24 h (<b>a</b>) and 48 h (<b>b</b>) of incubation. Magnification: 2000× (larger photo) and 5000× (smaller photo).</p>
Full article ">Figure 4
<p>Total cell numbers (CFU/mL) from mono-species and dual-species biofilms of <span class="html-italic">C. auris</span> and other <span class="html-italic">Candida</span> strains: (<b>a</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. albicans</span>, (<b>b</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. glabrata</span>, (<b>c</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. tropicalis</span>, and (<b>d</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. krusei</span>. The analyses were performed at 6, 24, and 48 h at 37 °C. At each time point of biofilm maturation, the CFU/mL of <span class="html-italic">Candida</span> from the mono-species and dual-species biofilms were compared with the CFU/mL of <span class="html-italic">C. auris</span> from the mono-species culture (black columns, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001) by Student’s <span class="html-italic">t</span>-test. Data are the means ± SD of experiments performed in of three duplicates and repeated two times.</p>
Full article ">Figure 5
<p>Survival curve of <span class="html-italic">G. mellonella</span> larvae infected with <span class="html-italic">Candida</span> strains: (<b>a</b>) larvae infected with single infections; (<b>b</b>) larvae infected with <span class="html-italic">C. auris</span> (single infection) and compared with larvae infected with <span class="html-italic">C. auris</span> and <span class="html-italic">C. albicans;</span> (<b>c</b>) <span class="html-italic">C. auris</span> and <span class="html-italic">C. tropicalis</span>; (<b>d</b>) <span class="html-italic">C. auris</span> and <span class="html-italic">C. glabrata</span>; and (<b>e</b>) <span class="html-italic">C. auris</span> and <span class="html-italic">C. krusei</span> (mixed infections). (<b>f</b>) Representative photo of <span class="html-italic">G. mellonella</span> larvae from a Petri dish culture.</p>
Full article ">Figure 6
<p>Total cell numbers (CFU/mL) of <span class="html-italic">Candida</span> species in <span class="html-italic">G. mellonella</span> hemolymph at various time points of dual-species experimental infections: (<b>a</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. albicans</span>, (<b>b</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. glabrata</span>, (<b>c</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. tropicalis</span>, and (<b>d</b>) <span class="html-italic">C. auris</span> with <span class="html-italic">C. krusei.</span> At each time point of infection, the CFU/mL of <span class="html-italic">Candida</span> from mono-species and dual-species infections were compared with the CFU/mL of <span class="html-italic">C. auris</span> from mono-species infections (* <span class="html-italic">p</span> &lt; 0.05) using the Student’s <span class="html-italic">t</span>-test. Data are the means ± SD of experiments performed in three repeats.</p>
Full article ">
16 pages, 5083 KiB  
Article
Integrating Bacteriocins and Biofilm-Degrading Enzymes to Eliminate L. monocytogenes Persistence
by John A. Renye, Chin-Yi Chen, Amanda Miller, Joe Lee, Adam Oest, Kevin J. Lynn, Samantha M. Felton, Manita Guragain, Peggy M. Tomasula, Bryan W. Berger and Joseph Capobianco
Int. J. Mol. Sci. 2025, 26(1), 399; https://doi.org/10.3390/ijms26010399 - 5 Jan 2025
Viewed by 765
Abstract
Listeria monocytogenes is a Gram-positive bacterium causing listeriosis, a severe infection responsible for significant morbidity and mortality globally. Its persistence on food processing surfaces via biofilm formation presents a major challenge, as conventional sanitizers and antimicrobials exhibit limited efficacy against biofilm-embedded cells. This [...] Read more.
Listeria monocytogenes is a Gram-positive bacterium causing listeriosis, a severe infection responsible for significant morbidity and mortality globally. Its persistence on food processing surfaces via biofilm formation presents a major challenge, as conventional sanitizers and antimicrobials exhibit limited efficacy against biofilm-embedded cells. This study investigates a novel approach combining an engineered polysaccharide-degrading enzyme (CAase) with a bacteriocin (thermophilin 110) produced by Streptococcus thermophilus. Laboratory assays evaluated the effectiveness of this combination in disrupting biofilms and inactivating L. monocytogenes on various surfaces. The results demonstrated that CAase effectively disrupts biofilm structures, while thermophilin 110 significantly reduces bacterial growth and viability. The preliminary trials indicate a dual-action approach offers a potential alternative to conventional treatments, enhancing food safety by effectively controlling Listeria biofilms in food processing environments. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Images of <span class="html-italic">L. monocytogenes</span> biofilms following treatment. The images depict mature biofilms following overnight treatment as follows: Control—PBS, CAase—0.1 mg/mL CAase, thermophilin 110–1000 AU/mL thermophilin 110, Mixture—a mixture of 0.1 mg/mL CAase and 1000 AU/mL thermophilin 110.</p>
Full article ">Figure 2
<p>Crystal violet assay results. Biofilm formation on microtiter plates as quantified by absorbance at 590 nm after crystal violet staining as described in <a href="#sec4dot7-ijms-26-00399" class="html-sec">Section 4.7</a>. Adsorption at 590 nm is plotted on the <span class="html-italic">y</span>-axis and the treatments are presented on the <span class="html-italic">x</span>-axis. The bars represent mean absorbance and error bars were constructed using 1 standard deviation from the mean of 3 biological replicates, each with 3 technical replicates. Groups not connected by the same letter were determined to be significantly different (<span class="html-italic">p</span> &lt; 0.05) by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Microscopy image analysis of CAase and thermophilin 110 effects on planktonic Listeria monocytogenes growth. (<b>A</b>) The normalized Background Corrected Absorption (BCAN) is plotted on the <span class="html-italic">y</span>-axis against time in hours on the <span class="html-italic">x</span>-axis. Analyzed images were recorded every 30 min. over the course of 25 h. The points represent an average of 3 experimental replicates, each containing 3 technical replicates (n = 9), and the error bars represent the standard deviation. (<b>B</b>) The area under each of the growth curves was calculated using Simpson’s method. The bars represent the average area (n = 9), while the error bars represent the standard deviation. The data were grouped by the composition of the growth media and level of inoculation. Within group comparisons were conducted using Student’s <span class="html-italic">t</span>-test and treatments not connected by the same letter were determined to be significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>oCelloScope images from media. Images from the oCelloScope that are grouped in rows by treatment and in columns by the time of measurement. The images were collected for the highest level of inoculation.</p>
Full article ">Figure 5
<p>oCelloScope images from PBS. The image displays images from the oCelloScope that are grouped in rows by treatment and in columns by the time of measurement. The images were collected for the highest level of inoculation.</p>
Full article ">Figure 6
<p>SEM images. <span class="html-italic">Listeria monocytogenes</span> cells at 50,000× magnification following 24 h treatment under the following conditions: Control (untreated), Thermophilin 110 (1000 AU/mL), CAase (0.1 mg/mL), and CAase + Thermophilin 110 (0.1 mg/mL CAase and 1000 AU/mL thermophilin 110). The scale bar represents 1.0 µm.</p>
Full article ">
13 pages, 2175 KiB  
Article
Genetic Determinants and Biofilm Properties Useful in Estimation of UTI Pathogenicity of the Escherichia coli Strains Isolated from Free-Living Birds
by Bartosz Rybak, Tomasz Jarzembowski, Agnieszka Daca, Beata Krawczyk and Lidia Piechowicz
Antibiotics 2025, 14(1), 32; https://doi.org/10.3390/antibiotics14010032 - 3 Jan 2025
Viewed by 596
Abstract
Background/Objectives: According to the One Health concept, wild birds can be indicators of ecosystem pollution and disease incidence. Escherichia coli strains are widespread worldwide, but there are still few reports on the association of human infections with a potential reservoir of highly [...] Read more.
Background/Objectives: According to the One Health concept, wild birds can be indicators of ecosystem pollution and disease incidence. Escherichia coli strains are widespread worldwide, but there are still few reports on the association of human infections with a potential reservoir of highly pathogenic human strains in wild birds. Fecal E. coli with uropathogenic potential (UPEC) can be transmitted between birds and humans and may be a risk factor for urinary tract infections (UTIs). Results: The results showed that above 50% of the isolates were grouped as highly pathogenic, according to Clermont phylogroup classification. Such strains were found to be stronger biofilm producers, with a higher adherence of monocytes than low pathogenic. However, the highest cytotoxicity was observed for strains described as aquatic environmental. Convergence of the results of the analysis of monocyte activation by E. coli strains and the ability to form biofilm by individual phylogroups of the strains tested was demonstrated. Genetic determinants of the uropathogenicity of E. coli (UPEC) correlate with the evidence of strain pathogenicity during monocyte activation in in vitro assays. Methods: In this study, we assessed the virulence potential of environmental strains isolated from wild waterfowl using genetic analysis (Clermont phylogroup classification) and phenotypic methods, including analysis of the human monocyte response to biofilm formation. The estimation of the ability to form biofilms was tested using crystal violet, and the pathogenic potential of strains by monocyte activation assay including changes in morphology, adhesion and cytotoxicity. Conclusions: In conclusion, the virulence of E. coli strains isolated from free-living birds is significant, and they can be considered environmental reservoirs of pathogenic strains. According to our observations, they can be responsible for the dissemination of uropathogenic strains among humans. Full article
(This article belongs to the Special Issue A One Health Approach to Antimicrobial Resistance)
Show Figures

Figure 1

Figure 1
<p>Drug susceptibility phenotypes of the tested <span class="html-italic">E. coli</span> strains (phylogroups by Clearmont—low pathogenic l: A; aquatic environmental: B1; highly pathogenic: B2, D).</p>
Full article ">Figure 2
<p>Results of the determination of the ability to form a biofilm with the use of crystal violet at the temperature of 37 °C (phylogroups by Clearmont—low pathogenic: A; aquatic environmental: B1; highly pathogenic: B2, D).</p>
Full article ">Figure 3
<p>Changes in the relative size of monocytes after the exposition to biofilm formed by tested strains. Vertical bars represent standard error (phylogroups by Clearmont—low pathogenic: A; aquatic environmental: B1; highly pathogenic: B2, D).</p>
Full article ">Figure 4
<p>Comparison of adherence of monocytes to bacterial biofilm formed by strains recognized as low pathogenic, environmental or highly pathogenic by phylogenetic analysis (ANOVA). Vertical bars represent standard error (phylogroups by Clearmont—low pathogenic: A; aquatic environmental: B1; highly pathogenic: B2, D).</p>
Full article ">Figure 5
<p>Cytotoxicity of bacterial biofilm formed by the strains recognized as low pathogenic, environmental or highly pathogenic by phylogenetic analysis. The value was estimated by monocyte cell membrane permeability after the exposition to the biofilm and measured as an increase of red fluorescence after PI staining. Vertical bars represent standard error (phylogroups by Clearmont: low pathogenic: A; aquatic environmental: B1; highly pathogenic: B2, D).</p>
Full article ">
10 pages, 535 KiB  
Article
Next-Generation Dental Materials: Exploring Bacterial Biofilm Formation on 3D-Printable Resin-Based Composites
by Emerson Koji Uehara, Gustavo Castro de Lima, Janaina de Cassia Orlandi Sardi, Luciene Cristina de Figueiredo, Jamil Awad Shibli, Thabet Asbi, Doron Haim and José Augusto Rodrigues
J. Funct. Biomater. 2025, 16(1), 12; https://doi.org/10.3390/jfb16010012 - 3 Jan 2025
Viewed by 521
Abstract
This study evaluated the microbial growth profile of subgingival multispecies biofilm on 3D-printable resin-based composites (PRBCs). A 96-well cell plate cultivated a 39-species biofilm associated with periodontitis over 7 days. Cylindrical specimens with 12 mm high and 3 mm diameters were prepared by [...] Read more.
This study evaluated the microbial growth profile of subgingival multispecies biofilm on 3D-printable resin-based composites (PRBCs). A 96-well cell plate cultivated a 39-species biofilm associated with periodontitis over 7 days. Cylindrical specimens with 12 mm high and 3 mm diameters were prepared by the PRBC group (Cosmos Temp-Yller; Prizma 3D Bio Crown; Prizma 3D Bio Prov) and an acrylic resin as control. Further, these specimens were immersed in the well plate to allow biofilm formation. After growing for 7 days, the metabolic biofilm activity was evaluated by colorimetric assay and the microbial profile by DNA-DNA hybridization. Kruskal–Wallis and Mann–Whitney tests evaluated each bacteria count and complex group. A greater biofilm formation was observed on PRBC groups than on acrylic resin. The microbiological profile of PRBC was associated with a less pathogenic biofilm, with an absence of a red complex. Acrylic resin showed low biofilm growth, but the biofilm profile was related to periodontal disease, characterized by red-complex bacteria. The selection of PRBC may contribute more effectively to maintaining periodontal health than acrylic resin. Full article
(This article belongs to the Special Issue Feature Papers in Dental Biomaterials (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Pie charts describing the total counts of microbial complexes (×10<sup>5</sup>) and percentage detected by Checkerboard DNA-DNA hybridization. The size of the pie charts represents the proportion of total bacterial counts.</p>
Full article ">
17 pages, 5886 KiB  
Article
Interference of Celastrol with Cell Wall Synthesis and Biofilm Formation in Staphylococcus epidermidis
by Leandro de León Guerra, Nayely Padilla Montaño and Laila Moujir
Antibiotics 2025, 14(1), 26; https://doi.org/10.3390/antibiotics14010026 - 3 Jan 2025
Viewed by 486
Abstract
Background: The emergence of antibiotic-resistant bacteria, including Staphylococcus epidermidis, underscores the need for novel antimicrobial agents. Celastrol, a natural compound derived from the plants of the Celastraceae family, has demonstrated promising antibacterial and antibiofilm properties against various pathogens. Objectives: This study [...] Read more.
Background: The emergence of antibiotic-resistant bacteria, including Staphylococcus epidermidis, underscores the need for novel antimicrobial agents. Celastrol, a natural compound derived from the plants of the Celastraceae family, has demonstrated promising antibacterial and antibiofilm properties against various pathogens. Objectives: This study aims to evaluate the antibacterial effects, mechanism of action, and antibiofilm activity of celastrol against S. epidermidis, an emerging opportunistic pathogen. Methods: To investigate the mechanism of action of celastrol, its antibacterial activity was evaluated by determining the time–kill curves, assessing macromolecular synthesis, and analysing its impact on the stability and functionality of the bacterial cell membrane. Additionally, its effect on biofilm formation and disruption was examined. Results: Celastrol exhibited significant antibacterial activity with a minimal inhibitory concentration (MIC) of 0.31 μg/mL and minimal bactericidal concentration (MBC) of 15 μg/mL, which is superior to conventional antibiotics used as control. Time–kill assays revealed a concentration-dependent bactericidal effect, with a shift from bacteriostatic activity at lower concentrations to bactericidal and lytic effect at higher concentrations. Celastrol inhibited cell wall biosynthesis by blocking the incorporation of N-acetylglucosamine (NAG) into peptidoglycan. In contrast, the cytoplasmic membrane was only affected at higher concentrations of the compound or after prolonged exposure times. Additionally, celastrol was able to disrupt biofilm formation at concentrations of 0.9 μg/mL and to eradicate pre-formed biofilms at 7.5 μg/mL in S. epidermidis. Conclusions: Celastrol exhibits significant antibacterial and antibiofilm activities against S. epidermidis, with a primary action on cell wall synthesis. Its efficacy in disrupting the formation of biofilms and pre-formed biofilms suggests its potential as a therapeutic agent for infections caused by biofilm-forming S. epidermidis resistant to conventional treatments. Full article
(This article belongs to the Special Issue Antimicrobial and Antibiofilm Activity by Natural Compounds)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chemical structure of celastrol.</p>
Full article ">Figure 2
<p>Killing curves of <span class="html-italic">S. epidermidis</span> in presence of celastrol or reference antibiotics. Untreated cell were used as a control. Compounds were added in lag-phase (<b>A</b>) and log-phase of growth after 4 h of preincubation (<b>B</b>). Error bars represent the standard deviation (SD) with <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 3
<p>Effect of celastrol (15 μg/mL) on different inoculum sizes of <span class="html-italic">S. epidermidis</span> (ranging from 10<sup>3</sup> to 10<sup>8</sup> CFU/mL) at lag-phase of growth. Log<sub>10</sub> of CFU counts (<b>A</b>) and optical density of the cultures (<b>B</b>). Error bars represent the standard deviation (SD) with <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 4
<p>Effect of celastrol at 15 μg/mL in the incorporation of radiolabeled precursors [6-<sup>3</sup>H] thymidine, [5-<sup>3</sup>H] uridine, [4,5-<sup>3</sup>H] leucine and N-Acetyl-D-[1-<sup>14</sup>C] glucosamine for the synthesis of DNA, RNA, protein and cell wall, respectively, in <span class="html-italic">S. epidermidis</span>. Data are expressed as percentage (%) of precursors incorporated compared to controls without drugs but with the maximum proportion of DMSO (100% of incorporation). Red line indicates 50% of inhibition. Error bars represent the standard deviation (SD) with <span class="html-italic">n</span> = 3. Different letters above bars mean significant differences between treated cultures within each given time point (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA; Tukey’s test).</p>
Full article ">Figure 5
<p>Effect of celastrol at 15 μg/mL in the uptake of radiolabeled precursors [6-<sup>3</sup>H] thymidine, [5-<sup>3</sup>H] uridine, [4,5-<sup>3</sup>H] leucine and N-Acetyl-D-[1-<sup>14</sup>C] glucosamine for the synthesis of DNA, RNA, protein and cell wall, respectively, in <span class="html-italic">S. epidermidis</span>. Data are expressed as percentage (%) of precursors uptaked compared to controls without drugs but with the maximum proportion of DMSO (100% of uptake). Red line indicates 50% of inhibition. Error bars represent the standard deviation (SD) with <span class="html-italic">n</span> = 3. Different letters above bars mean significant differences between treated cultures within each given time point (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA; Tukey’s test).</p>
Full article ">Figure 6
<p>Epifluorescence microscopy images of <span class="html-italic">S. epidermidis</span> stained with propidium iodide and Syto 9 after treatment with celastrol at 15 μg/mL for 60 min (<b>A</b>) and 120 min (<b>B</b>) or 30 μg/mL for 60 min (<b>C</b>) and 120 min (<b>D</b>).</p>
Full article ">Figure 7
<p>Effect of celastrol on the release of cytoplasmic components to the extracellular environment in <span class="html-italic">S. epidermidis.</span> Material absorbing at 260 nm (<b>A</b>) and 280 nm (<b>B</b>), and potassium release (<b>D</b>). Cell cultures treated with clofoctol served as positive control, while untreated cultures were used as negative control. Optical density at 435 nm (wavelength of maximal absorbance of celastrol) of cell suspension supernatant of <span class="html-italic">S. epidermidis</span> in saline buffer containing celastrol at 15 μg/mL (<b>C</b>). Error bars represent the standard deviation (SD) with <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 8
<p>Transmission Electron Microscopy (TEM) of <span class="html-italic">S. epidermidis</span> cells treated with celastrol at 15 µg/mL for 1 h (<b>A</b>,<b>B</b>). Cell cultures without drugs but containing the maximum DMSO concentration were used as controls (<b>C</b>). Arrows indicate the presence of mesosome-like structures at the division septum site.</p>
Full article ">Figure 9
<p>Effect of celastrol to inhibit biofilm formation or eradicate pre-formed biofilm of <span class="html-italic">S. epidermidis</span>. The percentage of inhibition or eradication is shown relative to the untreated control (not displayed), which represents 0% inhibition or eradication. The error bars represent standard deviation from the mean value. Error bars represent the standard deviation (SD) with <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 10
<p>Representative Scanning Electron Microscopy (SEM) images of <span class="html-italic">S. epidermidis</span> biofilm. (<b>A</b>,<b>B</b>) show untreated control cells with early biofilm formation after 24 h of culture. (<b>C</b>,<b>D</b>) illustrate the anti-adherence activity of celastrol (0.94 μg/mL) added at the beginning of the culture, resulting in dispersed cells and the inhibition of biofilm formation after 24 h. (<b>E</b>,<b>F</b>) depict the effect of celastrol (7.5 μg/mL) after 24 h of treatment on a pre-formed biofilm, showing disruption and eradication of typical cell aggregates observed in untreated cultures.</p>
Full article ">
19 pages, 3641 KiB  
Article
In Situ Aqueous Spice Extract-Based Antifungal Lock Strategy for Salvage of Foley’s Catheter Biofouled with Candida albicans Biofilm Gel
by Bindu Sadanandan, Vaniyamparambath Vijayalakshmi, Kalidas Shetty, Adithya Rathish, Harshala Shivkumar, Malavika Gundreddy, Nikhil Kumar Kagganti Narendra and Nethra Machamada Devaiah
Gels 2025, 11(1), 23; https://doi.org/10.3390/gels11010023 - 1 Jan 2025
Viewed by 534
Abstract
Candida forms a gel-like biofilm in the Foley’s catheter (FC) causing tenacious biofouling and severe urinary tract infections (UTIs). For the first time, a spice extract-based antifungal lock therapy (ALT) has been developed to inhibit the Candida albicans gel matrix in FC. Aqueous [...] Read more.
Candida forms a gel-like biofilm in the Foley’s catheter (FC) causing tenacious biofouling and severe urinary tract infections (UTIs). For the first time, a spice extract-based antifungal lock therapy (ALT) has been developed to inhibit the Candida albicans gel matrix in FC. Aqueous extracts of garlic, clove, and Indian gooseberry were used as ALT lock solutions and tested against biofilm-forming multidrug-resistant clinical isolates of C. albicans. Reduction in the gel matrices formation in the catheter was confirmed by Point inoculation, MTT assay, CFU, and SEM analysis at 12 and 24 h of incubation. Garlic was effective in controlling both C. albicans M207 and C. albicans S470; however, clove and gooseberry effectively controlled the latter. As evidenced by CFU assay, there were 82.85% and 99.68% reductions in the growth of C. albicans M207 and S470, respectively, at 24 h of incubation. SEM revealed a switch from the biofilm to the yeast mode and a drastic reduction in cell numbers, with mostly clumped or lysed cells. The study will provide an impetus to the development of novel spice extract-based ALT, reducing the selection pressure on the pathogen and lowering antimicrobial resistance. Further research in this area has the potential to leverage clinical applications. Full article
(This article belongs to the Special Issue Gels for Biomedical Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Panel 1: Point inoculation of the catheter sections at 12 h of incubation for (a) control, (b) garlic, (c) clove, and (d) gooseberry extracts. (A) <span class="html-italic">C. albicans</span> M207 center, (B) <span class="html-italic">C. albicans</span> M207 periphery, (C) <span class="html-italic">C. albicans</span> S470 centre, (D) <span class="html-italic">C. albicans</span> S470 periphery. Panel 2: Point inoculation of the catheter sections at 24 h of incubation for (a) control, (b) garlic, (c) clove, and (d) gooseberry extracts. (A) <span class="html-italic">C. albicans</span> M207 center, (B) <span class="html-italic">C. albicans</span> M207 periphery, (C) <span class="html-italic">C. albicans</span> S470 centre, (D) <span class="html-italic">C. albicans</span> S470 periphery.</p>
Full article ">Figure 1 Cont.
<p>Panel 1: Point inoculation of the catheter sections at 12 h of incubation for (a) control, (b) garlic, (c) clove, and (d) gooseberry extracts. (A) <span class="html-italic">C. albicans</span> M207 center, (B) <span class="html-italic">C. albicans</span> M207 periphery, (C) <span class="html-italic">C. albicans</span> S470 centre, (D) <span class="html-italic">C. albicans</span> S470 periphery. Panel 2: Point inoculation of the catheter sections at 24 h of incubation for (a) control, (b) garlic, (c) clove, and (d) gooseberry extracts. (A) <span class="html-italic">C. albicans</span> M207 center, (B) <span class="html-italic">C. albicans</span> M207 periphery, (C) <span class="html-italic">C. albicans</span> S470 centre, (D) <span class="html-italic">C. albicans</span> S470 periphery.</p>
Full article ">Figure 2
<p>Panel 1: MTT assay of center and periphery regions of the catheter having <span class="html-italic">C. albicans</span> M207 and <span class="html-italic">C. albicans</span> S470 grown for 12 h and treated with garlic, gooseberry, and clove. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01. The asterisk indicates a significant difference with respect to the control. Panel 2: MTT assay of center and periphery regions of catheter having <span class="html-italic">C. albicans</span> M207 and <span class="html-italic">C. albicans</span> S470 grown for 24 h and treated with garlic, gooseberry, and clove. **** <span class="html-italic">p</span> ≤ 0.0001. The asterisk indicates a significant difference with respect to the control.</p>
Full article ">Figure 3
<p>Panel 1: CFUs of (A) <span class="html-italic">C. albicans</span> M207 control treated with (B) garlic extract at 12 and 24 h of incubation. (a) 12 h neat, (b) 12 h 10<sup>−1</sup> dilution, (c) 24 h neat, (d) 24 h 10<sup>−1</sup> dilution. Panel 2: CFUs of (A) <span class="html-italic">C. albicans</span> S470 control treated with (B) garlic, (C) gooseberry, and (D) clove extracts at 12 and 24 h of incubation. (a) 12 h neat, (b) 12 h 10<sup>−1</sup> dilution, (c) 24 h neat, (d) 24 h 10<sup>−1</sup> dilution.</p>
Full article ">Figure 4
<p>SEM analysis of the longitudinal section of the catheter. Panel 1: Blank catheter. Panel 2: (A) <span class="html-italic">C. albicans</span> M207 at 12 h, (B) <span class="html-italic">C. albicans</span> S470 at 12 h, (C) <span class="html-italic">C. albicans</span> M207 at 24 h and (D) <span class="html-italic">C. albicans</span> S470 at 24 h. (a) Control, (b) garlic-treated, (c) gooseberry-treated, (d) clove-treated.</p>
Full article ">Figure 5
<p>The proposed mechanism of action of allicin, eugenol/ellagic acid, and gallic acid of garlic (pink arrow), clove (black arrow), and gooseberry (green arrow), respectively, on <span class="html-italic">Candida</span> sp. Allicin causes cellular damage, affects the sodium–potassium pump, affects lipid synthesis, causes lipid peroxidation, inactivates thiol peptide (glutathione) and proteins (glutathione peroxidase, glutathione reductase, coenzyme A) that act as innate antioxidants, leading to oxidative stress, thereby also triggering ROS generation, damages mitochondria, reduces succinate dehydrogenase, produces ROS, which, in turn, cause cytochrome C release, caspase activation and apoptosis, inhibits ECE1 virulence factor/candidalysin, and inactivates quorum-sensing genes. Eugenol/ellagic acid causes cellular damage, releases cytochrome C, leading to caspase activation and apoptosis, produces ROS, releases nucleic acid and proteins, inhibits ECE1 virulence factor/candidalysin, and forms DNA adducts. Gallic acid causes cellular damage, leading to cytoplasmic leakage, damage to nucleic acid and proteins, and inhibits RNA synthesis. Classes of conventional antimycotics (azole, allylamine, echinocandins, polyenes, the miscellaneous class—griseofulvin, and nucleotide analogues) and their binding sites are also depicted in the figure.</p>
Full article ">
Back to TopTop