[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
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

Search Results (1,475)

Search Parameters:
Keywords = drug transfer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1183 KiB  
Review
The Global Challenge of Campylobacter: Antimicrobial Resistance and Emerging Intervention Strategies
by Zubeiru Bukari, Toyin Emmanuel, Jude Woodward, Richard Ferguson, Martha Ezughara, Nikhil Darga and Bruno Silvester Lopes
Trop. Med. Infect. Dis. 2025, 10(1), 25; https://doi.org/10.3390/tropicalmed10010025 - 16 Jan 2025
Viewed by 368
Abstract
Antimicrobial resistance (AMR) in Campylobacter species, particularly C. jejuni and C. coli, poses a significant public health threat. These bacteria, which are commonly found in livestock, poultry, companion animals, and wildlife, are the leading causes of foodborne illnesses, often transmitted through contaminated [...] Read more.
Antimicrobial resistance (AMR) in Campylobacter species, particularly C. jejuni and C. coli, poses a significant public health threat. These bacteria, which are commonly found in livestock, poultry, companion animals, and wildlife, are the leading causes of foodborne illnesses, often transmitted through contaminated poultry. Extensive exposure to antibiotics in human and veterinary medicine creates selection pressure, driving resistance through mechanisms such as point mutations, horizontal gene transfer, and efflux pumps. Resistance to fluoroquinolones, macrolides, and tetracyclines complicates treatment and increases the risk of severe infections. Drug-resistant Campylobacter is transmitted to humans via contaminated food, water, and direct contact with animals, highlighting its zoonotic potential. Addressing this challenge requires effective interventions. Pre-harvest strategies like biosecurity and immune-based methods reduce bacterial loads on farms, while post-harvest measures, including carcass decontamination and freezing, limit contamination. Emerging approaches, such as bacteriocins and natural antimicrobials, offer chemical-free alternatives. Integrated, multidisciplinary interventions across the food chain are essential to mitigate AMR transmission and enhance food safety. Sustainable agricultural practices, antimicrobial stewardship, and innovative solutions are critical to curbing Campylobacter resistance and protecting global public health. Our review examines the dynamics of antimicrobial resistance in Campylobacter and presents current strategies to mitigate Campylobacter-related AMR, offering valuable insights for antimicrobial control in the poultry industry. Full article
Show Figures

Figure 1

Figure 1
<p>Transmission pathways in <span class="html-italic">Campylobacter</span>.</p>
Full article ">Figure 2
<p>Intervention strategies used to tackle <span class="html-italic">Campylobacter</span>.</p>
Full article ">
10 pages, 867 KiB  
Article
Comparative Analyses of Antiviral Potencies of Second-Generation Integrase Strand Transfer Inhibitors (INSTIs) and the Developmental Compound 4d Against a Panel of Integrase Quadruple Mutants
by Steven J. Smith, Xue Zhi Zhao, Stephen H. Hughes and Terrence R. Burke
Viruses 2025, 17(1), 121; https://doi.org/10.3390/v17010121 - 16 Jan 2025
Viewed by 238
Abstract
Second-generation integrase strand transfer inhibitors (INSTIs) are strongly recommended for people living with HIV-1 (PLWH). The emergence of resistance to second-generation INSTIs has been infrequent and has not yet been a major issue in high-income countries. However, the delayed rollouts of these INSTIs [...] Read more.
Second-generation integrase strand transfer inhibitors (INSTIs) are strongly recommended for people living with HIV-1 (PLWH). The emergence of resistance to second-generation INSTIs has been infrequent and has not yet been a major issue in high-income countries. However, the delayed rollouts of these INSTIs in low- to middle-income countries during the COVID-19 pandemic combined with increased transmission of drug-resistant mutants worldwide are leading to an increase in INSTI resistance. Herein, we evaluated the antiviral potencies of our lead developmental INSTI 4d and the second-generation INSTIs dolutegravir (DTG), bictegravir (BIC), and cabotegravir (CAB) against a panel of IN quadruple mutants. The mutations are centered around G140S/Q148H, including positions L74, E92, and T97 combined with E138A/K/G140S/Q148H. All of the tested INSTIs lose potency against these IN quadruple mutants compared with the wild-type IN. In single-round infection assays, compound 4d retained higher antiviral potencies (EC50 values) than second-generation INSTIs against a subset of quadruple mutants. These findings may advance understanding of mechanisms that contribute to resistance and, in so doing, facilitate development of new INSTIs with improved antiviral profiles. Full article
(This article belongs to the Collection Efficacy and Safety of Antiviral Therapy)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of INSTIs used in this study. Chemical structures of FDA-approved second-generation INSTIs and compound 4d are depicted. Atoms shown in red represent the chelating motifs, while the linker group is in purple and the halogenated benzyl moiety in blue.</p>
Full article ">Figure 2
<p>IN residues that undergo mutation during INSTI resistance development. BIC (in purple) in the active site of the HIV-1 intasome (IN is in light blue, while viral DNA end is shown in gray; PDB: 6PUW). Mutations to IN residues depicted in orange are frequently selected by INSTIs and are often found in INSTI-resistant IN double and triple mutants. IN resistance mutations also arise in the residues shown in green. Mutations at these positions can be a part of the IN quadruple mutants that have been analyzed in this study. All residues and nucleotides are labeled in black.</p>
Full article ">
32 pages, 3650 KiB  
Review
Mechanistic Insights into the Anticancer Potential of Methoxyflavones Analogs: A Review
by Mohammad Aidiel, Maisarah Abdul Mutalib, Rajesh Ramasamy, Nik Nasihah Nik Ramli, Shirley Gee Hoon Tang and Siti Hajar Adam
Molecules 2025, 30(2), 346; https://doi.org/10.3390/molecules30020346 - 16 Jan 2025
Viewed by 244
Abstract
2-phenylchromen-4-one, commonly known as flavone, plays multifaceted roles in biological response that can be abundantly present in natural sources. The methoxy group in naturally occurring flavones promotes cytotoxic activity in various cancer cell lines by targeting protein markers, in facilitating ligand–protein binding mechanisms [...] Read more.
2-phenylchromen-4-one, commonly known as flavone, plays multifaceted roles in biological response that can be abundantly present in natural sources. The methoxy group in naturally occurring flavones promotes cytotoxic activity in various cancer cell lines by targeting protein markers, in facilitating ligand–protein binding mechanisms and activating cascading downstream signaling pathways leading to cell death. However, the lipophilic nature of these analogs is a key concern as it impacts drug membrane transfer. While lipophilicity is crucial for drug efficacy, the excessive lipophilic effects in flavonoids can reduce water solubility and hinder drug transport to target sites. Recent in vitro studies suggest that the incorporation of polar hydroxyl groups which can form hydrogen bonds and stabilize free radicals may help overcome the challenges associated with methoxy groups while maintaining their essential lipophilic properties. Naturally coexisting with methoxyflavones, this review explores the synergistic role of hydroxy and methoxy moieties through hydrogen bonding capacity in maximizing cytotoxicity against cancer cell lines. The physicochemical analysis revealed the potential intramolecular interaction and favorable electron delocalization region between both moieties to improve cytotoxicity levels. Together, the analysis provides a useful strategy for the structure–activity relationship (SAR) of flavonoid analogs in distinct protein markers, suggesting optimal functional group positioning to achieve balanced lipophilicity, effective hydrogen bonding, and simultaneously minimized steric hindrance in targeting specific cancer cell types. Full article
Show Figures

Figure 1

Figure 1
<p>Skeleton structure of flavone analogs.</p>
Full article ">Figure 2
<p>The influence of the position of both functional groups in ring B on the IC<sub>50</sub> value in MCF-7 in 72 h treatment duration [<a href="#B47-molecules-30-00346" class="html-bibr">47</a>,<a href="#B48-molecules-30-00346" class="html-bibr">48</a>,<a href="#B49-molecules-30-00346" class="html-bibr">49</a>]. The presence of both methoxy and hydroxy moieties on neighboring positions (C3′ and C4′) is a crucial factor for a stronger IC<sub>50</sub> of 5-OH methoxyflavones analogs. The C6,7,8-OCH<sub>3</sub> plays a crucial role in stabilizing the lipophilic capacity of ring A flavones. Given the presence of C6 and C7-OCH<sub>3</sub>, the reduction of single C8-OCH<sub>3</sub> will not significantly affect the IC<sub>50</sub> of these scaffolds. For xanthomicrol, the absence of methoxy adjacent to the C4′-OH was a single factor in the expected weak cytotoxicity profile.</p>
Full article ">Figure 3
<p>Schematic diagram on anticancer activity of methoxyflavones scaffold. The C7 position plays a key role in the cytotoxic activity on cancer cell lines.</p>
Full article ">Figure 4
<p>Lewis’s resonance structures illustrating electron delocalization in (<b>A</b>) C3′, (<b>B</b>) C4′, and (<b>C</b>) C5 free radicals. Ring AC and BC are not conjugated. According to a 2013 study, a strong IHB between C5-OH and the C4 oxygen atom of the carbonyl group facilitates the formation of IHBs involving (<b>D</b>) C4′-OH and (<b>E</b>) C3′-OH with an adjacent methoxy groups.</p>
Full article ">Figure 5
<p>Ring B dihydroxylated moieties highlight the impact of C5′ scaffold composition to the cytotoxic effect on HCC1954 breast cancer cell lines [<a href="#B64-molecules-30-00346" class="html-bibr">64</a>]. The presence of C5-OCH<sub>3</sub> shows enhanced cytotoxicity similar to the C5-OH methoxyflavones scaffolds, influenced by C5′-OH. As summarized in <a href="#molecules-30-00346-f003" class="html-fig">Figure 3</a>, an excessive methoxylated effect on ring B yielded a negative IC<sub>50</sub> result on cancer cell lines.</p>
Full article ">Figure 6
<p>SAR analysis of methoxyflavones analogs and their cytotoxic effects on prostate cancer cell lines. The arrow directions show the transformation from one compound to another, detailing the specific chemical changes like the addition or removal of hydroxyl (-OH) and methoxy (-OCH<sub>3</sub>) groups.</p>
Full article ">Figure 7
<p>SAR analysis on cytotoxicity activity of methoxyflavones analogs on HCT116 colon cancer cell lines between 24 and 72 h treatment duration [<a href="#B87-molecules-30-00346" class="html-bibr">87</a>,<a href="#B88-molecules-30-00346" class="html-bibr">88</a>,<a href="#B89-molecules-30-00346" class="html-bibr">89</a>]. The molecular structures of various methoxyflavones and their respective impacts on cell viability.</p>
Full article ">Figure 8
<p>SAR analysis on cytotoxicity activities of 5-demethylnobiletin derivatives on HepG2 liver cancer cell lines in 24 h treatment [<a href="#B91-molecules-30-00346" class="html-bibr">91</a>]. Each derivative (1–4) shows a different substitution pattern on the ring B methoxyflavone structure. The IC<sub>50</sub> values adjacent to each structure illustrate the effect of these modifications on cytotoxic efficacy. Notably, derivative 2 with a hydroxyl group at position R<sub>3′</sub> shows the lowest IC<sub>50</sub> value of 41.37 µM, suggesting increased efficacy compared to other derivatives.</p>
Full article ">Figure 9
<p>SAR analysis on cytotoxicity activities of 5-demethylnobiletin, sinensetin, and sudachitin analogs on HepG2 colon cancer cell lines for 24 to 48 h treatment [<a href="#B87-molecules-30-00346" class="html-bibr">87</a>,<a href="#B91-molecules-30-00346" class="html-bibr">91</a>,<a href="#B93-molecules-30-00346" class="html-bibr">93</a>]. The arrows indicate structural transformations and their direct impact on cytotoxic efficacy, illustrating how specific modifications in the flavonoid skeleton influence cell death pathways in HepG2 cells.</p>
Full article ">
18 pages, 4252 KiB  
Article
Bilayer TiO2/Mo-BiVO4 Photoelectrocatalysts for Ibuprofen Degradation
by Martha Pylarinou, Elias Sakellis, Spiros Gardelis, Vassilis Psycharis, Marios G. Kostakis, Nikolaos S. Thomaidis and Vlassis Likodimos
Materials 2025, 18(2), 344; https://doi.org/10.3390/ma18020344 - 14 Jan 2025
Viewed by 336
Abstract
Heterojunction formation between BiVO4 nanomaterials and benchmark semiconductor photocatalysts has been keenly pursued as a promising approach to improve charge transport and charge separation via interfacial electron transfer for the photoelectrocatalytic degradation of recalcitrant pharmaceutical pollutants. In this work, a heterostructured TiO [...] Read more.
Heterojunction formation between BiVO4 nanomaterials and benchmark semiconductor photocatalysts has been keenly pursued as a promising approach to improve charge transport and charge separation via interfacial electron transfer for the photoelectrocatalytic degradation of recalcitrant pharmaceutical pollutants. In this work, a heterostructured TiO2/Mo-BiVO4 bilayer photoanode was fabricated by the deposition of a mesoporous TiO2 overlayer using the benchmark P25 titania catalyst on top of Mo-doped BiVO4 inverse opal films as the supporting layer, which intrinsically absorbs visible light below 490 nm, while offering improved charge transport. A porous P25/Mo-BiVO4 bilayer structure was produced from the densification of the inverse opal underlayer after post-thermal annealing, which was evaluated on photocurrent generation in aqueous electrolyte and the photoelectrocatalytic degradation of the refractory anti-inflammatory drug ibuprofen under back-side illumination by visible and UV–Vis light. Significantly enhanced photoelectrochemical performance on both photocurrent density and pharmaceutical degradation was achieved for the bilayer structure with respect to the additive effect of the constituent layers, which was related to the improved light harvesting arising from the backscattering by the mesoporous TiO2 layer in combination with the favorable charge transfer at the TiO2/Mo-BiVO4 interface. Full article
(This article belongs to the Special Issue Feature Papers in Materials Physics (2nd Edition))
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>a</b>) TEM image of the Mo-BiVO<sub>4</sub> PC underlayer and the corresponding (<b>b</b>) Bi, (<b>c</b>) V, (<b>d</b>) O, and (<b>e</b>) Mo EDX elemental maps. (<b>f</b>) Top view and (<b>g</b>) cross-section SEM image of the P25/Mo-BiVO<sub>4</sub> bilayer structure. (<b>h</b>) Cross-section SEM image of the bottom Mo-BiVO<sub>4</sub> layer.</p>
Full article ">Figure 2
<p>(<b>a</b>) XRD patterns of the P25/Mo-BiVO<sub>4</sub> bilayer in comparison to the individual Mo-BiVO<sub>4</sub> and P25 layer, the undoped BiVO<sub>4</sub> PC film, and the FTO substrate. Shaded bands indicate the 2θ regions where the variations of the (<b>b</b>) main (101) diffraction peak of the anatase TiO<sub>2</sub> phase of P25, and the splitting of the (<b>c</b>) (002)/(200) and (<b>d</b>) (240)/(042) diffraction peaks of the <span class="html-italic">ms</span> BiVO<sub>4</sub> phase, are shown in detail. Red solid lines in (<b>b</b>) depict the best fits of the (101) peaks to Voight profiles with full widths at half-maximum of 0.40 and 0.41 deg for the P25 and P25/Mo-BiVO<sub>4</sub> bilayer films.</p>
Full article ">Figure 3
<p>Raman spectra of the P25/Mo-BiVO<sub>4</sub> bilayer in comparison to the single BiVO<sub>4</sub>, Mo-BiVO<sub>4</sub> and P25 layers at (<b>a</b>) 785 and (<b>b</b>) 532 nm excitation wavelengths. Dashed boxes delineate the spectral regions that are shown in detail in the shaded insets. In the left inset for each laser excitation, the anatase E<sub>g</sub> modes of the P25 and P25/Mo-BiVO<sub>4</sub> films are compared after background removal at normalized intensity scale. Arrows indicate the weak shoulder that is associated with the Mo-O stretching vibration for the Mo-BiVO<sub>4</sub> layers.</p>
Full article ">Figure 4
<p>(<b>a</b>) Diffuse reflectance (DR%), (<b>b</b>) transmittance (T%) and the corresponding (<b>c</b>) Kubelka–Munk absorption, and (<b>d</b>) Tauc plots of the individual layers and the P25/Mo-BiVO<sub>4</sub> bilayers.</p>
Full article ">Figure 5
<p>Photocurrent density-potential (J-V) curves for the P25/Mo-BiVO<sub>4</sub> and Mo-BiVO<sub>4</sub>/P25 films in comparison to the individual layers and BiVO<sub>4</sub> reference under (<b>a</b>) visible and (<b>b</b>) UV–Vis back-side illumination in 0.5 M NaHCO<sub>3</sub> aqueous electrolyte. (<b>c</b>) IPCE spectra at 1.23 V vs. RHE potential for the P25/Mo-BiVO<sub>4</sub> bilayer with respect to the P25 and Mo-BiVO<sub>4</sub> constituent layers.</p>
Full article ">Figure 6
<p>Mott–Schottky plots for the P25/Mo-BiVO<sub>4</sub> bilayer in comparison to the Mo-BiVO<sub>4</sub> and P25 individual films under (<b>a</b>) dark, (<b>b</b>) visible, and (<b>c</b>) UV–Vis light illumination in 0.5 M NaHCO<sub>3</sub> aqueous electrolyte. (<b>d</b>) The derived values of <span class="html-italic">V</span><sub>fb</sub> and <span class="html-italic">N</span><sub>D</sub> and (<b>e</b>) tentative band alignment for the Mo-BiVO<sub>4</sub>/TiO<sub>2</sub> heterojunction before and after Fermi level equilibration.</p>
Full article ">Figure 7
<p>EIS Nyquist plots for the P25/Mo-BiVO<sub>4</sub> bilayer in comparison to Mo-BiVO<sub>4</sub> and P25 films under (<b>a</b>) dark, (<b>b</b>) visible, and (<b>c</b>) UV–Vis back-side illumination in 0.5 M NaHCO<sub>3</sub> aqueous electrolyte. Black lines denote the best fit curves to the Randles circuit shown in (<b>a</b>).</p>
Full article ">Figure 8
<p>(<b>a</b>,<b>b</b>) IBU photoelectrocatalytic degradation kinetics and (<b>c</b>) corresponding kinetic constants for the P25 films in 0.1 M NaHCO<sub>3</sub> supporting electrolyte at bias potentials of 0, +0.5, and +1.0 V vs. Ag/AgCl under UV–Vis illumination.</p>
Full article ">Figure 9
<p>(<b>a</b>) IBU absorbance spectra under UV–Vis illumination in the presence of P25/Mo-BiVO<sub>4</sub>. IBU photodegradation kinetics under (<b>b</b>,<b>c</b>) visible and (<b>d</b>,<b>e</b>) UV–Vis light as well as the corresponding (<b>f</b>) reaction rates for the individual constituents and the P25/Mo-BiVO<sub>4</sub> bilayer photoelectrodes at +1.0 V vs. Ag/AgCl.</p>
Full article ">Figure 10
<p>Absorbance spectra of (<b>a</b>) IBU and (<b>b</b>) the degradation product. HPLC chromatograms of IBU in the presence of P25/Mo-BiVO<sub>4</sub> films under UV–Vis light for (<b>c</b>) 0, (<b>d</b>) 1, (<b>e</b>) 2, and (<b>f</b>) 5 h.</p>
Full article ">Figure 11
<p>(<b>a</b>) Three consecutive IBU photoelectrocatalytic degradation tests using the same P25/Mo-BiVO<sub>4</sub> bilayer films under UV–Vis light at +1.0 V vs. Ag/AgCl. (<b>b</b>) Raman spectra of the bilayer film before and after the third photocatalytic run at 532 nm.</p>
Full article ">Scheme 1
<p>Fabrication process of P25/Mo-BiVO<sub>4</sub> bilayer films.</p>
Full article ">
22 pages, 4827 KiB  
Article
Synthesis of Anti-Inflammatory Drugs’ Chalcone Derivatives and a Study of Their Conformational Properties Through a Combination of Nuclear Magnetic Resonance Spectroscopy and Molecular Modeling
by Nikitas Georgiou, Andromachi Tzani, Kyriaki Vavougyiou, Christos Papadopoulos, Nikolaos Eleftheriadis, Primož Šket, Demeter Tzeli, Tuomas Niemi-Aro, Anastasia Detsi and Thomas Mavromoustakos
Pharmaceuticals 2025, 18(1), 88; https://doi.org/10.3390/ph18010088 - 13 Jan 2025
Viewed by 297
Abstract
Background: In this study, two chalcone analogs were synthesized through in silico and experimental methods, and their potential to inhibit the lipoxygenase enzyme, which plays a role in the inflammation pathway, was assessed. Specifically, this study is a continuation of previous research in [...] Read more.
Background: In this study, two chalcone analogs were synthesized through in silico and experimental methods, and their potential to inhibit the lipoxygenase enzyme, which plays a role in the inflammation pathway, was assessed. Specifically, this study is a continuation of previous research in which chalcone derivatives were synthesized and characterized. Objectives/Methods: In the current work, we present the re-synthesis of two chalcones, with a focus on their docking studies, NMR analysis, and dynamic simulations. The structure of each chalcone was elucidated through a combination of Nuclear Magnetic Resonance (NMR) and Density Functional Theory (DFT). The substituent effect on the absorption spectrum of the two chalcone derivatives was studied. Results: A “LOX–chalcone” complex, predicted by docking studies, was further examined using molecular dynamics (MD) simulations to evaluate the stability of the complex. After fully characterizing the “LOX–chalcone” complexes in silico, the atomic details of each chalcone’s interaction with LOX-1 and 5-LOX were revealed through Saturation Transfer Difference (STD) NMR (Nuclear Magnetic Resonance). Finally, their selectivity profile was investigated against human 15-LOX-1 and general Lipoxidase activity. Conclusions: The in silico methods suggest that chalcones could be promising lead compounds for drug designs targeting the LOX enzyme. Full article
(This article belongs to the Special Issue Chalcones: Structure, Function, and Applications)
Show Figures

Figure 1

Figure 1
<p><sup>1</sup>H NMR spectra in DMSO of (<b>a</b>) <b>1</b> and (<b>b</b>) <b>2</b>. The spectra were recorded in DMSO-d<sub>6</sub> on a Bruker AC 850 MHz spectrometer at 25 °C.</p>
Full article ">Figure 2
<p>The lowest-energy arrangements for (<b>a</b>) <b>1</b> and (<b>b</b>) <b>2</b>, along with their spatial correlations, which dictate their lowest-energy configurations; distances in Å for the DFT (BP86/def2-SVP) methodology (red is oxygen, green is chlorium).</p>
Full article ">Figure 3
<p>Interactions of <b>1</b> with (<b>a</b>) 5-LOX and (<b>b</b>) 1-LOX in 2D (<b>above</b>) and 3D (<b>bottom</b>) formats.</p>
Full article ">Figure 4
<p>Interactions of <b>2</b> with (<b>a</b>) 5-LOX and (<b>b</b>) 1-LOX in 2D (<b>above</b>) and 3D (<b>bottom</b>) formats.</p>
Full article ">Figure 5
<p>Interactions of <b>1</b> with (<b>a</b>) 15-LOX and <b>2</b> with (<b>b</b>) 15-LOX in 2D (<b>above</b>) and 3D (<b>bottom</b>) formats.</p>
Full article ">Figure 6
<p>RMSD values for the protein (5-LOX) (depicted in green) and ligands 1 (<b>a</b>) and 2 (<b>b</b>) (depicted in magenta) were calculated over a 200 ns simulation period, with the initial docking pose serving as the reference structure (upper image).</p>
Full article ">Figure 7
<p>RMSD values for the protein (LOX-1) (depicted in green) and ligands 1 (<b>a</b>) and 2 (<b>b</b>) (depicted in magenta) were calculated over a 200 ns simulation period, with the initial docking pose serving as the reference structure (upper image).</p>
Full article ">Figure 8
<p>RMSD values for the protein (15-LOX) (depicted in green) and ligands 1 (<b>a</b>) and 2 (<b>b</b>) (depicted in magenta) were calculated over a 200 ns simulation period, with the initial docking pose serving as the reference structure (upper image).</p>
Full article ">Figure 9
<p>(Top) The STD-NMR reference spectrum of <b>1</b> (1000 mM) in the presence of 5-LOX protein (1 mM) at a 1:1000 ratio with respect to <b>1</b> was obtained in a potassium phosphate buffer at pH 7.2 and 600 μL D<sub>2</sub>O using an 850 MHz NMR spectrometer at 25 °C. (Middle) The STD-NMR reference spectrum of <b>1</b> (1000 mM) in the presence of the LOX-1 protein (1 mM) at a 1:100 ratio with respect to <b>1</b> was obtained in potassium phosphate buffer at pH 7.2 and 600 μL D<sub>2</sub>O using an 850 MHz NMR spectrometer at 25 °C. (Bottom) <sup>1</sup>H spectra of <b>1</b> in DMSO, obtained using an 850 MHz NMR spectrometer at 25 °C.</p>
Full article ">Figure 10
<p>(Top) The STD-NMR reference spectrum of <b>2</b> (1000 mM) in the presence of 5-LOX protein (1 mM) at a 1:1000 ratio with respect to <b>2</b> was obtained in potassium phosphate buffer at pH 7.2 and 600 μL D<sub>2</sub>O using an 850 MHz NMR spectrometer at 25 °C. (Middle) The STD-NMR reference spectrum of <b>2</b> (1000 mM) in the presence of LOX-1 protein (1 mM) at a 1:100 ratio with respect to <b>2</b> was obtained in potassium phosphate buffer at pH 7.2 and 600 μL D<sub>2</sub>O using an 850 MHz NMR spectrometer at 25 °C. (Bottom) <sup>1</sup>H spectra of <b>2</b> in DMSO, obtained using an 850 MHz NMR spectrometer at 25 °C.</p>
Full article ">Figure 11
<p>Absorption spectra of the free calculated compounds and those encapsulated in 5-LOX and in LOX-1, obtained using the B3LYP/def2-SVP method. Emission spectra of the free calculated compounds obtained using the B3LYP/def2-SVP method. The geometries of the encapsulated complexes’ structures, obtained via MD calculations.</p>
Full article ">Figure 12
<p>Frontier molecular orbitals (HOMO on the left and LUMO on the right) involved in the S<sub>1</sub> absorption peaks in the (<b>a</b>) free 1, (<b>b</b>) 1 complexed with 5-LOX, (<b>c</b>) 1 complexed with LOX-1 after MD, (<b>d</b>) free 2, (<b>e</b>) 2 complexed with 5-LOX, and (<b>f</b>) 2 complexed with LOX-1, and frontier molecular orbitals involved in the S1→S0 fluorescence peaks in the (<b>g</b>) free 1 and (<b>h</b>) free 2.</p>
Full article ">Figure 13
<p>Compounds tested (at 100 μΜ) against human 15-LOX-1 and Lipoxidase enzymes. Positive control (absence of potential inhibitor) was set to 100% (dash line = 85%). The averages and standard deviations of the individual measurements are presented.</p>
Full article ">Scheme 1
<p>General structure and numbering scheme of chalcones.</p>
Full article ">Scheme 2
<p>Structures of chalcones <b>1</b> and <b>2</b>.</p>
Full article ">
14 pages, 901 KiB  
Article
Mapping Antimicrobial Resistance in Staphylococcus epidermidis Isolates from Subclinical Mastitis in Danish Dairy Cows
by Desiree Corvera Kløve, Mikael Lenz Strube, Peter M. H. Heegaard and Lærke Boye Astrup
Antibiotics 2025, 14(1), 67; https://doi.org/10.3390/antibiotics14010067 - 10 Jan 2025
Viewed by 458
Abstract
Background/Objectives: Although Staphylococcus epidermidis is a key cause of subclinical mastitis in Danish dairy cows, its sensitivity to antimicrobials remains unexplored. Here, we analyzed sixty S. epidermidis isolates derived from 42 dairy cows across six conventional dairy herds in Denmark. Methods: Phenotypic resistance [...] Read more.
Background/Objectives: Although Staphylococcus epidermidis is a key cause of subclinical mastitis in Danish dairy cows, its sensitivity to antimicrobials remains unexplored. Here, we analyzed sixty S. epidermidis isolates derived from 42 dairy cows across six conventional dairy herds in Denmark. Methods: Phenotypic resistance was measured by antimicrobial susceptibility testing and minimum inhibitory concentration (MIC) analysis, and genotypic resistance was examined through whole-genome sequencing and identification of antimicrobial resistance genes (ARGs). Correspondence between phenotypic and genotypic resistance was then evaluated by Cohen’s kappa statistics. Furthermore, the presence of plasmid replicon genes and the strain diversity among the S. epidermidis isolates was investigated to associate these findings with the observed AMR patterns. Results: Results showed that 30/60 isolates (50.0%) were resistant to penicillin phenotypically, while 35/60 (58.3%) were positive for a corresponding blaZ gene (κ = 0.83, p < 0.01). A fosB gene, encoding fosfomycin resistance, was detected in all 60/60 isolates (100.0%), but fosfomycin resistance was not analyzed phenotypically. Based on MIC analysis, 3/60 isolates (5.0%) were multi-drug resistant, showing resistance towards penicillin, erythromycin, and tetracycline. However, in 11/60 genomes (18.3%), ARGs encoding resistance towards ≥3 antimicrobial classes (e.g., beta-lactams, phosphonic acid, tetracyclines, aminoglycosides, macrolides, lincosamides, and fusidane) were detected. Eleven different ARGs were detected among the 60 isolates in total. No methicillin-resistant Staphylococcus epidermidis (MRSE) were recorded. Results further showed that each herd had one primary sequence type (ST) and resistance profile associated with it, and plasmid-mediated horizontal gene transfer of ARGs was indicated This study underscores the importance of routine resistance surveillance and species-specific diagnoses to improve treatment outcomes and ensure prudent use of antimicrobials. Full article
(This article belongs to the Special Issue Antimicrobial Resistance of Pathogens Isolated from Bovine Mastitis)
Show Figures

Figure 1

Figure 1
<p>Core-genome phylogeny of 60 <span class="html-italic">S. epidermidis</span> isolates from SCM in Danish dairy cows. The isolates are named according to isolate number (SE1 to SE60), herd number (H1 to H6), and cow number (C1 to C7). The isolates are further named “a” or “b” in cases of cows with <span class="html-italic">S. epidermidis</span> in two quarters of the udder. The phylogenic tree is annotated with ARGs (dark green for positive) and plasmid replicon genes (light green for positive). <sup>a</sup> rep5b_4_SAP106B007(SAP106B). <sup>b</sup> rep5b_5_SAP108B006(SAP108B). <sup>c</sup> rep7a_4_repD(pK214), <sup>d</sup> rep7a_16_repC(Cassette), <sup>e</sup> rep13_6_rep(pLNU1), <sup>f</sup> rep20_11_repA(VRSAp). ST is presented in blue color scheme (ND = ST not determined).</p>
Full article ">
8 pages, 873 KiB  
Article
Antimicrobial Susceptibility of Glaesserella parasuis to Macrolides and Characterization of erm(T)-Carrying Mobile Elements on Chromosome
by Peng Zhang, Changmin Li, Shuna Shang, Ting Huang, Junqi Liu, Qianwen Ge, Xiaoping Liao, Liangxing Fang and Yang Yu
Animals 2025, 15(2), 164; https://doi.org/10.3390/ani15020164 - 10 Jan 2025
Viewed by 302
Abstract
Glaesserella parasuis is the etiological agent of Glässer’s disease, which causes high morbidity and mortality in pigs worldwide. Macrolide resistance poses an urgent threat to their treatment, as macrolides are widely used for preventing and treating G. parasuis infections. Here, we determined the [...] Read more.
Glaesserella parasuis is the etiological agent of Glässer’s disease, which causes high morbidity and mortality in pigs worldwide. Macrolide resistance poses an urgent threat to their treatment, as macrolides are widely used for preventing and treating G. parasuis infections. Here, we determined the susceptibilities to five macrolides and characterized the genetic markers of macrolide resistance. The antimicrobial susceptibility of 117 G. parasuis isolates to erythromycin, tulathromycin, gamithromycin, tylosin, and tilmicosin was evaluated using broth microdilution method. Erythromycin-resistant isolates were sequenced using whole-genome sequencing. Further analysis of these sequences revealed the genetic basis of macrolide resistance in G. parasuis. Our results show that most G. parasuis isolates remained susceptible to the macrolide drugs. For commonly used agents (e.g., tylosin and tilmicosin), elevated minimum inhibitory concentrations (MICs) were observed, whereas for the newer macrolides (e.g., tulathromycin and gamithromycin), the MICs remained almost unchanged. The macrolide resistance gene erm(T) and the A2059G mutation in 23S rRNA were detected in the current study. To the best of our knowledge, integrative and conjugative element (ICE)-borne erm(T) in G. parasuis is reported for the first time in this study. Taken together, these results provide insights into the susceptibility of G. parasuis to macrolides. The presence of erm(T) on ICEs may facilitate its transfer, reducing the effectiveness of macrolide treatment. Full article
(This article belongs to the Section Pigs)
Show Figures

Figure 1

Figure 1
<p>Putative ICEs identified in this study: ICE<span class="html-italic">Hpa1</span>-like01 and ICE<span class="html-italic">Hpa1</span>-like02. AMR gene, relaxase, transposase, integrase, ICE-related function sequences, and hypothetical proteins are indicated by colors according to the legend. Scale bar represents 10 kb pairs. AMR, antimicrobial resistance; ICEs, integrative and conjugative elements.</p>
Full article ">
13 pages, 3142 KiB  
Article
Advanced Large-Stokes-Shift Fluorescent Probe for the Detection of Biothiols: Facilitating Accurate Indirect Measurement of β-Lactamases
by Likun Liu, Dongling Yan, Yukun Ma, Peng Hou, Pengfei Qi, Xue Zhang, Yitong Liu and Song Chen
Int. J. Mol. Sci. 2025, 26(2), 525; https://doi.org/10.3390/ijms26020525 - 9 Jan 2025
Viewed by 284
Abstract
A novel fluorescent probe, Bibc-DNBS, based on the combination of the PET (photoinduced electron transfer) and ESIPT (excited-state intramolecular proton transfer) mechanisms, was designed and synthesized. Bibc-DNBS exhibited a Stokes shift of 172 nm in the fluorescence detection field. In addition, the probe [...] Read more.
A novel fluorescent probe, Bibc-DNBS, based on the combination of the PET (photoinduced electron transfer) and ESIPT (excited-state intramolecular proton transfer) mechanisms, was designed and synthesized. Bibc-DNBS exhibited a Stokes shift of 172 nm in the fluorescence detection field. In addition, the probe exhibited good performance in key parameters in bioassays such as sensitivity, specificity, and response time. Based on these properties, Bibc-DNBS successfully monitored the biothiol levels in live cells and zebrafish models, providing an effective analytical tool for real-time monitoring of biothiols. More importantly, Bibc-DNBS could be useful for indirectly detecting β-lactamases. Bibc-DNBS(3-(1H-benzo[d]imidazol-2-yl)-4′-cyano-[1,1′-biphenyl]-4-yl2,4-dinitrobenzenesulfonate) facilitated the screening of β-lactamase inhibitors, using tazobactam and clavulanic acid as model compounds, with respective semi-inhibitory concentration values of 31.32 μM and 2.26 μM, respectively. It might also be applied to distinguish sensitive strain Staphylococcus aureus ATCC 29213 and drug-resistant strain Enterobacter cloacae ATCC 13047, which could provide strong support for the clinical application of antibiotics and the development of new drugs. Full article
(This article belongs to the Special Issue Luminescent Dyes as Tools for Biological and Medical Applications)
Show Figures

Figure 1

Figure 1
<p>Fluorescence changes in the fluorescent probe Bibc-DNBS (10.0 μM) interacting with 0.0–100.0 μM Cys (cysteine, <b>A</b>)/Hcy (homocysteine, <b>B</b>)/GSH (glutathione, <b>C</b>). (<b>1</b>) Fluorescence spectrogram; (<b>2</b>) fluorescence intensity versus Cys/Hcy/GSH concentration; (<b>3</b>) linear fit of fluorescence intensity to (0.0–10.0 μM) Cys/Hcy/GSH. Inset: cuvette image of probe Bibc-DNBS before (a) and after reaction (b) with Cys taken under a handheld UV lamp (λ<sub>ex</sub> = 365 nm), scale bar 0.5 cm.</p>
Full article ">Figure 2
<p>(<b>A</b>) Detection of thiols in HepG2 cells by probe Bibc-DNBS confocal imaging. (<b>a1</b>–<b>a3</b>) Control: HepG2 cells not incubated with the probe Bibc-DNBS. (<b>b1</b>–<b>b3</b>) HepG2 cells were incubated with the probe Bibc-DNBS (10.0 μM) for 30 min. (<b>c1</b>–<b>c3</b>) HepG2 cells were first stimulated with NEM (N-ethylmaleimide, 1.0 mM) for 30 min, followed by incubation with Bibc-DNBS (10.0 μM) for an additional 30 min. (<b>d1</b>–<b>d3</b>) NEM-HepG2 cells were treated with 100.0 μM Cys, (<b>e1</b>–<b>e3</b>) 100.0 μM Hcy, (<b>f1</b>–<b>f3</b>) 100.0 μM GSH for 30 min, and incubated with 10.0 μM Bibc-DNBS for additional 30 min, respectively. (<b>B</b>) Fluorescence intensities in panels (<b>a</b>–<b>f</b>). n = 3; error bars are ±SD. Statistical analysis was performed with a one-way ANOVA. Among them, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. Scale bar 20 μm.</p>
Full article ">Figure 3
<p>(<b>A</b>) Imaging experiments for detection of thiols in zebrafish. (<b>a1</b>–<b>a3</b>) Control: zebrafish not incubated with the probe Bibc-DNBS. (<b>b1</b>–<b>b3</b>) Zebrafish was incubated with Bibc-DNBS (10.0 μM) for 30 min. (<b>c1</b>–<b>c3</b>) Zebrafish was pretreated with NEM (1.0 mM) for 30 min, then incubated with probe Bibc-DNBS (10.0 μM) for another 30 min. (<b>B</b>) Fluorescence intensities in panels (<b>a</b>–<b>c</b>). n = 3; error bars are ±SD. Statistical analysis was performed with a one-way ANOVA. Among them, *** <span class="html-italic">p</span> &lt; 0.001. Scale bar 100 μm.</p>
Full article ">Figure 4
<p>(<b>A</b>) Fluorescence spectra of Bibc-DNBS upon addition of different concentrations of β-lactamase (0–0.16 U/mL) containing 250.0 μM cefazolin sodium. Inset: cuvette image of probe Bibc-DNBS before (a) and after reaction (b) with β-lactamase taken under a handheld UV lamp (λ<sub>ex</sub> = 365 nm), scale bar 0.5 cm. (<b>B</b>) With the concentration of β-lactamase being (0.0–0.16 U/mL), the fluorescence signal intensity scattering diagram of Bibc-DNBS (10.0 μM) at 462 nm. (<b>C</b>) Linear diagram of fluorescence signal intensity of probe and β-lactamase (0.000–0.010 U/mL).</p>
Full article ">Figure 5
<p>(<b>A</b>) HPLC chromatogram of the reaction of cefazolin sodium with β-lactamase: (red) MMT standard, (green) cefazolin sodium, (blue) cefazolin sodium reacted with beta-lactamase for 0.5 h, and (purple) cefazolin sodium reacted with beta-lactamase for 1 h. (<b>B</b>) HPLC chromatogram of Bibc-DNBS reacting with β-lactamase: (orange) Bibc-OH, (cyan) Bibc-DNBS, (yellow) Bibc-DNBS reacted with MMT standard, and (pink) cefazolin sodium reacted with β-lactamase and Bibc-DNBS. Conditions: eluent H<sub>2</sub>O/CH<sub>3</sub>CN (<span class="html-italic">v</span>/<span class="html-italic">v</span>, 90/10); flow rate 1.0 mL/min; temperature 30 °C; sample injection volume 20.0 μL.</p>
Full article ">Figure 6
<p>(<b>A</b>) The fluorescence intensity of Bibc-DNBS with or without β-lactamase was recorded by enzyme-labeling instrument when different antibiotics were used as substrates. (<b>B</b>) Inhibition circle test (1–6 cefazolin sodium, cefotiam sodium, cefonicid sodium, blank reagents, ceftriaxone sodium, and cefamandole sodium); scale bar 1 cm.</p>
Full article ">Scheme 1
<p>(<b>A</b>) Synthetic route to Bibc-DNBS. (<b>B</b>) Mechanism of thiol recognition and the indirect detection of β-lactamase by Bibc-DNBS.</p>
Full article ">
14 pages, 6588 KiB  
Article
Sustainable Corrosion Inhibitors from Pharmaceutical Wastes: Advancing Energy-Efficient Chemistry with Green Solutions
by Narasimha Raghavendra, Sharanappa Chapi, Murugendrappa M. V., Małgorzata Pawlak and Mohammad Reza Saeb
Energies 2025, 18(2), 224; https://doi.org/10.3390/en18020224 - 7 Jan 2025
Viewed by 452
Abstract
Pharmaceutical waste is a type of bio-waste inevitably generated by the pharmaceutical industry, often due to regulatory changes, product deterioration, or expiration. However, their collection and valorization can be approached from a sustainable perspective, offering potential energy-efficient solutions. In this work, the expired [...] Read more.
Pharmaceutical waste is a type of bio-waste inevitably generated by the pharmaceutical industry, often due to regulatory changes, product deterioration, or expiration. However, their collection and valorization can be approached from a sustainable perspective, offering potential energy-efficient solutions. In this work, the expired Eslicarbazepine acetate drug (ESLD) was utilized as a sustainable anticorrosive agent against mild steel in a 3 M HCl wash solution. Experimental tests combined with theoretical Density Functional Theory (DFT) and Monte Carlo (MC) simulations revealed the corrosion inhibition potential of ESLD. The gasometrical results revealed a high inhibition efficiency rate of 98% upon increases in concentration of expired ESLD from 0.25 to 1.00 mg·L−1, whereas hydrogen gas evolution decreased to 0.7 mL. An impedance investigation evidenced the pivotal role of charge transfer in reducing the disintegration process. As per DFT computations and MC simulation, electron-rich elements in the expired ESLD were key in controlling the dissolution through the adsorption process. Contact angle studies revealed that the increment in the contact angle from 61° to 80° in the presence of expired ESLD validates the chemical, electrochemical, and computational results. This approach not only mitigates pharmaceutical pollution, but also exemplifies the integration of green chemistry principles into corrosion protection, contributing to energy-efficient and sustainable industrial practices. Full article
(This article belongs to the Section B: Energy and Environment)
Show Figures

Figure 1

Figure 1
<p>Interactive chemical structure model (Ball and Stick) and chemical structure depiction of ESLD.</p>
Full article ">Figure 2
<p>Tafel plots with and without an inhibitor for MS in 3 M HCl solution.</p>
Full article ">Figure 3
<p>Nyquist curves for the MS exposed to the blank solution and acidic electrolytes with four distinct expired ESLD concentrations and equivalent circuits used in the present investigation (<a href="#energies-18-00224-f003" class="html-fig">Figure 3</a>).</p>
Full article ">Figure 4
<p>The relationship between the concentration of inhibitor and protection efficiency at different exposure periods.</p>
Full article ">Figure 5
<p>SEM images of (<b>a</b>) without and (<b>b</b>) with 1 mg/L of expired ESL drug at room temperature.</p>
Full article ">Figure 6
<p>Water contact angle measurement. (<b>a</b>) Bare and (<b>b</b>) inhibited system.</p>
Full article ">Figure 7
<p>DFT results: (<b>a</b>) HOMO, (<b>b</b>) LUMO, and (<b>c</b>) density mapping of an expired drug.</p>
Full article ">Figure 8
<p>Images of the different views of the adsorption of expired ESLD molecules on the Fe surface in an aqueous environment.</p>
Full article ">Figure 8 Cont.
<p>Images of the different views of the adsorption of expired ESLD molecules on the Fe surface in an aqueous environment.</p>
Full article ">Figure 9
<p>Diagram interpretation of the adsorption of expired ESLD on MS in acidic environments.</p>
Full article ">
21 pages, 9923 KiB  
Article
Trust Region Policy Learning for Adaptive Drug Infusion with Communication Networks in Hypertensive Patients
by Mai The Vu, Seong Han Kim, Ha Le Nhu Ngoc Thanh, Majid Roohi and Tuan Hai Nguyen
Mathematics 2025, 13(1), 136; https://doi.org/10.3390/math13010136 - 1 Jan 2025
Viewed by 425
Abstract
In the field of biomedical engineering, the issue of drug delivery constitutes a multifaceted and demanding endeavor for healthcare professionals. The intravenous administration of pharmacological agents to patients and the normalization of average arterial blood pressure (AABP) to desired thresholds represents a prevalent [...] Read more.
In the field of biomedical engineering, the issue of drug delivery constitutes a multifaceted and demanding endeavor for healthcare professionals. The intravenous administration of pharmacological agents to patients and the normalization of average arterial blood pressure (AABP) to desired thresholds represents a prevalent approach employed within clinical settings. The automated closed-loop infusion of vasoactive drugs for the purpose of modulating blood pressure (BP) in patients suffering from acute hypertension has been the focus of rigorous investigation in recent years. In previous works where model-based and fuzzy controllers are used to control AABP, model-based controllers rely on the precise mathematical model, while fuzzy controllers entail complexity due to rule sets. To overcome these challenges, this paper presents an adaptive closed-loop drug delivery system to control AABP by adjusting the infusion rate, as well as a communication time delay (CTD) for analyzing the wireless connectivity and interruption in transferring feedback data as a new insight. Firstly, a nonlinear backstepping controller (NBC) is developed to control AABP by continuously adjusting vasoactive drugs using real-time feedback. Secondly, a model-free deep reinforcement learning (MF-DRL) algorithm is integrated into the NBC to adjust dynamically the coefficients of the controller. Besides the various analyses such as normal condition (without CTD strategy), stability, and hybrid noise, a CTD analysis is implemented to illustrate the functionality of the system in a wireless manner and interruption in real-time feedback data. Full article
(This article belongs to the Special Issue Artificial Intelligence for Biomedical Applications)
Show Figures

Figure 1

Figure 1
<p>The overall structure of the closed-loop AABP mechanism.</p>
Full article ">Figure 2
<p>Dynamic model of the AABP system with NBC and the reference input: (<b>a</b>) Dynamic model, (<b>b</b>) reference input.</p>
Full article ">Figure 3
<p>The structure of the policy and value networks.</p>
Full article ">Figure 4
<p>The flowchart of the TRPO algorithm.</p>
Full article ">Figure 5
<p>The overall structure of tuning NBC’s coefficients by TRPO with CTD technique.</p>
Full article ">Figure 6
<p>The functionality of the system: (<b>a</b>) Normal, (<b>b</b>) fixed CTD, (<b>c</b>) random CTD.</p>
Full article ">Figure 7
<p>The outcomes of the developed strategy when <math display="inline"><semantics> <mrow> <mi>P</mi> </mrow> </semantics></math> changes, (<b>a</b>–<b>c</b>) is for AABP, and (<b>d</b>–<b>f</b>) is for drug dosage.</p>
Full article ">Figure 7 Cont.
<p>The outcomes of the developed strategy when <math display="inline"><semantics> <mrow> <mi>P</mi> </mrow> </semantics></math> changes, (<b>a</b>–<b>c</b>) is for AABP, and (<b>d</b>–<b>f</b>) is for drug dosage.</p>
Full article ">Figure 8
<p>The outcomes of the developed strategy when <math display="inline"><semantics> <mrow> <msub> <mrow> <mi mathvariant="sans-serif">Χ</mi> </mrow> <mrow> <mi>i</mi> <mn>1</mn> </mrow> </msub> </mrow> </semantics></math> changes, (<b>a</b>–<b>c</b>) is for AABP, and (<b>d</b>–<b>f</b>) is for drug dosage.</p>
Full article ">Figure 9
<p>The results of the developed strategy when reference changes.</p>
Full article ">Figure 10
<p>The outputs of the various controllers under <math display="inline"><semantics> <mrow> <mi>P</mi> </mrow> </semantics></math> changes.</p>
Full article ">Figure 11
<p>The outputs of the various controllers under <math display="inline"><semantics> <mrow> <msub> <mrow> <mi mathvariant="sans-serif">Χ</mi> </mrow> <mrow> <mi>i</mi> <mn>1</mn> </mrow> </msub> </mrow> </semantics></math> changes.</p>
Full article ">Figure 12
<p>RMS results of the created strategy when there is a hybrid noise.</p>
Full article ">
24 pages, 2226 KiB  
Review
Liposomal Formulations: A Recent Update
by Surendra S. Agrawal, Vrinda Baliga and Vaishali Y. Londhe
Pharmaceutics 2025, 17(1), 36; https://doi.org/10.3390/pharmaceutics17010036 - 30 Dec 2024
Viewed by 1148
Abstract
Liposome-based drug delivery technologies have showed potential in enhancing medication safety and efficacy. Innovative drug loading and release mechanisms highlighted in this review of next-generation liposomal formulations. Due to poor drug release kinetics and loading capacity, conventional liposomes have limited clinical use. Scientists [...] Read more.
Liposome-based drug delivery technologies have showed potential in enhancing medication safety and efficacy. Innovative drug loading and release mechanisms highlighted in this review of next-generation liposomal formulations. Due to poor drug release kinetics and loading capacity, conventional liposomes have limited clinical use. Scientists have developed new liposomal carrier medication release control and encapsulation methods to address these limits. Drug encapsulation can be optimized by creating lipid compositions that match a drug’s charge and hydrophobicity. By selecting lipids and adding co-solvents or surfactants, scientists have increased drug loading in liposomal formulations while maintaining stability. Nanotechnology has also created multifunctional liposomes with triggered release and personalized drug delivery. Surface modification methods like PEGylation and ligand conjugation can direct liposomes to disease regions, improving therapeutic efficacy and reducing off-target effects. In addition to drug loading, researchers have focused on spatiotemporal modulation of liposomal carrier medication release. Stimuli-responsive liposomes release drugs in response to bodily signals. Liposomes can be pH- or temperature-sensitive. To improve therapeutic efficacy and reduce systemic toxicity, researchers added stimuli-responsive components to liposomal membranes to precisely control drug release kinetics. Advanced drug delivery technologies like magnetic targeting and ultrasound. Pro Drug, RNA Liposomes approach may improve liposomal medication administration. Magnetic targeting helps liposomes aggregate at illness sites and improves drug delivery, whereas ultrasound-mediated drug release facilitates on-demand release of encapsulated medicines. This review also covers recent preclinical and clinical research showing the therapeutic promise of next-generation liposomal formulations for cancer, infectious diseases, neurological disorders and inflammatory disorders. The transfer of these innovative liposomal formulations from lab to clinical practice involves key difficulties such scalability, manufacturing difficulty, and regulatory limits. Full article
(This article belongs to the Special Issue Advanced Liposomes for Drug Delivery, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Novel methods for delivery of liposomes.</p>
Full article ">Figure 2
<p>Normalized ATR-FTIR spectra of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine/ampholytic molecular switch liposomes (absorption region of asymmetric stretching vibrations of methylene groups) under various conditions: (<b>a</b>) liposomes at pH 5.0 (blue), at pH 8.4 (red dashed line), and converted from pH 5.0 to 8.4 (green); (<b>b</b>) liposomes at pH 5.0 (blue dashed line), at pH 8.4 (red line), and converted from pH 8.4 to pH 5.0 (yellow). Total lipid concentration 5 mg/mL, αAMS = 0.1, 22 °C. SEM images of <span class="html-italic">C. crispus</span>, <span class="html-italic">G. gracilis</span>, and <span class="html-italic">G. Corneum</span> 3D gels. Reprinted/adapted with permission from Ref. [<a href="#B82-pharmaceutics-17-00036" class="html-bibr">82</a>].</p>
Full article ">Figure 3
<p>Live imaging of PC-3 cells under LTSL-BPNF therapy Reprinted/adapted with permission from Ref. [<a href="#B91-pharmaceutics-17-00036" class="html-bibr">91</a>]. After seeding 96-well plates with 2 × 10<sup>4</sup> PC-3 cells each, the cells were allowed to proliferate for the entire night. The cells were exposed to a dose of 0.5 mM of LTSL-BPNFs for three hours the next day. The cells were then exposed to a 10 min laser irradiation with a power density of 1 W/cm<sup>2</sup>. The cells were then incubated for 30 min after the addition of 1 µM of DHE to the culture medium. After washing the cells with HBSS to remove any leftover fluorescent probe, the MuviCyte kit was used to take live cell pictures. The scale bar has a length of 500 μm. SEM images of <span class="html-italic">C. crispus</span>, <span class="html-italic">G. gracilis</span>, and <span class="html-italic">G. Corneum</span> 3D gels.</p>
Full article ">Figure 4
<p>Mechanism of enzyme-responsive liposomes.</p>
Full article ">
17 pages, 3929 KiB  
Article
Exploring Zinc C295 as a Dual HIV-1 Integrase Inhibitor: From Strand Transfer to 3′-Processing Suppression
by Sharif Karim Sayyed, Marzuqa Quraishi, D. S. Prabakaran, Balaji Chandrasekaran, Thiyagarajan Ramesh, Satish Kumar Rajasekharan, Chaitany Jayprakash Raorane, Tareeka Sonawane and Vinothkannan Ravichandran
Pharmaceuticals 2025, 18(1), 30; https://doi.org/10.3390/ph18010030 - 29 Dec 2024
Viewed by 634
Abstract
Background: The global AIDS pandemic highlights the urgent need for novel antiretroviral therapies (ART). In our previous work, Zinc C295 was identified as a potent HIV-1 integrase strand transfer (ST) inhibitor. This study explores its potential to also inhibit 3′-processing (3′P), thereby [...] Read more.
Background: The global AIDS pandemic highlights the urgent need for novel antiretroviral therapies (ART). In our previous work, Zinc C295 was identified as a potent HIV-1 integrase strand transfer (ST) inhibitor. This study explores its potential to also inhibit 3′-processing (3′P), thereby establishing its dual-targeting capability. Methods: The inhibitory activity of Zinc C295 against 3′P was evaluated using a modified in vitro assay adapted from our earlier ST inhibition studies. Molecular docking and molecular dynamics simulations were employed to analyse Zinc C295’s interactions with the 3′P allosteric site of HIV-1 integrase. Results: Zinc C295 demonstrated significant inhibition of HIV-1 integrase 3′P activity in in vitro assays (IC50 = 4.709 ± 0.97 µM). Computational analyses revealed key interactions of Zinc C295 within the enzyme’s allosteric site, providing insights into its dual inhibitory mechanism. Conclusions: Zinc C295’s dual inhibition of HIV-1 integrase ST and 3′P establishes it as a promising candidate for next-generation ART. Its dual-action mechanism may offer potential advantages in enhancing treatment efficacy and addressing drug resistance. Further studies are warranted to evaluate its therapeutic potential in clinical settings. Full article
(This article belongs to the Special Issue In Silico and In Vitro Screening of Small Molecule Inhibitors)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Percent inhibition of HIV-1 (<b>A</b>) strand transfer (ST) activity and (<b>B</b>) integrase 3′ processing (3′P) activity.</p>
Full article ">Figure 2
<p>Visualization of molecular docking interactions between BI-1001 and Zinc C295 with the HIV-1 IN catalytic core domain (CCD) allosteric site (PDB ID: 4DMN). The figure presents 3D representations of the docking interactions generated using PyMOL and 2D representations created with Maestro, along with the ligand structures drawn in Marvin: (<b>A</b>) BI-1001 (magenta), (<b>B</b>) Zinc C295 (green), and (<b>C</b>) the combined interactions of BI-1001 and Zinc C295 with HIV-1 IN. In the 2D interaction diagram, the residues involved in hydrogen bonds are indicated in blue, including Gln95, Thr124, and Thr125, while the residues engaged in hydrophobic interactions are shown in green, such as Trp132, Ala128, and Ala129. Key binding interactions are highlighted to illustrate the molecular interactions involved in the binding efficacy of both ligands.</p>
Full article ">Figure 3
<p>MD simulation analysis of the Zinc C295-4DMN complex over 100 ns. (<b>A</b>) <b>RMSD plot</b> showing the stability and fluctuations of the 4DMN protein and Zinc C295, with protein RMSD ranging from 1.5 to 4 Å, indicating overall structural stability. (<b>B</b>) <b>RMSF plot</b> identifying flexible regions, particularly around residue indices 60, 70, and 130, suggesting potential ligand interaction sites. (<b>C</b>) <b>Bar graph</b> illustrating the fraction of time key residues, including Ala133, Ala98, Tyr99, and Leu102, interacted with Zinc C295, highlighting hydrophobic contacts. (<b>D</b>) <b>Ligand–protein contact map</b> detailing interactions, including hydrogen bonds and water-mediated contacts, with residues such as Ala128 and Trp132, contributing to the stability of the Zinc C295-4DMN complex.</p>
Full article ">Figure 4
<p>Mechanism of HIV Integrase Inhibition. This schematic illustrates two types of integrase inhibitors: strand transfer inhibitors (INSTIs) (<b>left</b>) and allosteric integrase inhibitors (<b>right</b>). INSTIs inhibit the IN enzyme by preventing the joining of the target substrate DNA to the donor substrate DNA following the 3′ processing (3′P) step. In contrast, allosteric inhibitors act by blocking the 3′ processing step itself, thereby preventing IN from binding to DS DNA and interfering with the integration process.</p>
Full article ">Figure 5
<p>Schematic representation of HIV infection with inhibitory effects demonstrated by the proposed lead compounds against stages of IN (strand transfer and 3′ processing). Abbreviations: C295 is Zinc C295, ELV is Elvitegravir.</p>
Full article ">
16 pages, 8723 KiB  
Article
Heterologous Expression of a Potential ‘Paulownia fortunei’ MYB Factor Gene, PfMYB90, Improves Salt and Cold Tolerance in Arabidopsis
by Hongling Wang, Shizheng Shi, Guijie Luo, Ruifang Huang, Dezong Sui, Yunpeng Gao and Lei Wang
Plants 2025, 14(1), 24; https://doi.org/10.3390/plants14010024 - 25 Dec 2024
Viewed by 375
Abstract
The paulownia tree belongs to the Paulowniaceae family. Paulownia has strong vitality; has strong adaptability to harsh environmental conditions; and can be used as building raw material, as well as processing drugs and having other purposes. In the research field of MYB transcription [...] Read more.
The paulownia tree belongs to the Paulowniaceae family. Paulownia has strong vitality; has strong adaptability to harsh environmental conditions; and can be used as building raw material, as well as processing drugs and having other purposes. In the research field of MYB transcription factors of the paulownia tree, it is rare to discuss the resistance to abiotic stress. The research in this area has not received sufficient attention and depth, which also indicates an important potential direction for future research. In this study, we performed bioinformatics analysis of the stress-related gene PfMYB90, a potential transcription factor, and investigated its mechanism of action under salt and cold stresses. PfMYB90 was strongly expressed in the fully unfolded leaf and root of plants in both stress treatments. Transgenic PfMYB90 Arabidopsis plants had a greater survival rate under salt and cold stresses, and the degree of leaf damage was comparatively smaller, according to phenotypic observation and survival rate calculations. By measuring the corresponding physiological indexes after stress and detecting the expression levels of corresponding stress genes (AtNHX1, AtSOS1, AtSOS2, AtSOS3, AtCBF1, AtCBF3, AtCOR15a, AtRD29a), it was found that after PfMYB90 gene transfer, Arabidopsis showed strong tolerance to salt and cold stresses. This is consistent with the results mentioned above. This transgenic technology enables Arabidopsis to survive under adverse environmental conditions, allowing it to maintain a relatively stable growth state despite salt accumulation and cold stress. Therefore, PfMYB90 may be a key gene in the regulatory network of salt damage and cold damage, as well as one of the key transcription factors for Paulownia fortunei environmental conditions. Full article
Show Figures

Figure 1

Figure 1
<p>PfMYB90 protein multiple sequencing analysis. (<b>A</b>) Alignment of MYB sequence. (<b>B</b>) The evolution tree. Target proteins are represented by the red lines, while R-repeat structures are shown by the yellow and green boxes.</p>
Full article ">Figure 2
<p>Subcellular location of PfMYB90. There are four frames in the image: (<b>A</b>,<b>E</b>) NLS-mCherry, (<b>B</b>,<b>F</b>) GFP, (<b>C</b>,<b>G</b>) Bright, and (<b>D</b>,<b>H</b>) Merge. bar = 30 μm.</p>
Full article ">Figure 3
<p>Expression of <span class="html-italic">PfMYB90</span>. (<b>A</b>) <span class="html-italic">PfMYB90</span> expression in various organs. <span class="html-italic">PfMYB90</span> expression levels in young leaf as a reference. (<b>B</b>) Relative expression of <span class="html-italic">PfMYB90</span> under stresses in fully unfolded leaf and (<b>C</b>) root. The expression level of 0 h was set to 1 as the control (CK). An error bar (<span class="html-italic">n</span> = 3) represents the standard deviation. ** <span class="html-italic">p</span>-value ≤ 0.01 and * <span class="html-italic">p</span>-value ≤ 0.05.</p>
Full article ">Figure 4
<p>Transgenic-<span class="html-italic">PfMYB90</span> enhanced the salt tolerance of <span class="html-italic">Arabidopsis</span>. (<b>A</b>) <span class="html-italic">PfMYB90</span> expression in transgenic lines (L1-6), unloaded lines (UL), and wild type (WT) <span class="html-italic">Arabidopsis</span> plants. (<b>B</b>) Phenotypes of <span class="html-italic">Arabidopsis</span> WT, UL, L1, L3, and L5 after 0 d (Salt 0 d) and 8 d (Salt 8 d) of treatment with 100 mM NaCl. Bar is equal to 4 cm. (<b>C</b>) <span class="html-italic">Arabidopsis</span> survival rates at Salt 0 d and Salt 8 d. Applying WT as a guide. An error bar (<span class="html-italic">n</span> = 3) represents the standard deviation. ** <span class="html-italic">p</span>-value ≤ 0.01 and * <span class="html-italic">p</span>-value ≤ 0.05.</p>
Full article ">Figure 5
<p>Analysis of (<b>A</b>) SOD, (<b>B</b>) POD, (<b>C</b>) proline, (<b>D</b>) chlorophyll, (<b>E</b>) CAT, (<b>F</b>) and MDA in <span class="html-italic">Arabidopsis</span> at 0 d (Salt 0 d) and 8 d (Salt 8 d) of treatment with 100 mM NaCl. Using WT indicators as controls. The SD is represented by an error bar (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span>-value ≤ 0.01.</p>
Full article ">Figure 6
<p><span class="html-italic">Arabidopsis</span> salt-tolerance-related gene expression was detected by RT-qPCR. Relative expression levels of (<b>A</b>) <span class="html-italic">AtNHX1</span>, (<b>B</b>) <span class="html-italic">AtSOS1</span>, (<b>C</b>) <span class="html-italic">AtSOS2</span>, and (<b>D</b>) <span class="html-italic">AtSOS3</span> after 0 d (Salt 0 d) and 8 d (Salt 8 d) of treatment with 100 mM NaCl. Using WT as control. The SD is represented by an error bar (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span>-value ≤ 0.01.</p>
Full article ">Figure 7
<p><span class="html-italic">Arabidopsis</span> phenotypic alterations and survival rate under cold stress. (<b>A</b>) The <span class="html-italic">Arabidopsis</span> phenotype (Cold 0 h, 12 h, and recover), with a scale of 4 cm. (<b>B</b>) Survival rate of <span class="html-italic">Arabidopsis</span> under CK (Cold 0 h) and cold stress (Cold 12 h). The SD is represented by an error bar (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span>-value ≤ 0.01.</p>
Full article ">Figure 8
<p>Impact of the <span class="html-italic">PfMYB90</span> gene on the <span class="html-italic">Arabidopsis</span> cold tolerance index. WT was utilized as the control in (<b>A</b>) SOD, (<b>B</b>) POD, (<b>C</b>) proline, (<b>D</b>) chlorophyll, (<b>E</b>) CAT, and (<b>F</b>) MDA. There is an error bar (<span class="html-italic">n</span> = 3) that shows the SD. ** <span class="html-italic">p</span>-value ≤ 0.01.</p>
Full article ">Figure 9
<p>Expression levels of genes linked to cold stress in transgenic, UL, and WT <span class="html-italic">Arabidopsis</span> under cold stress conditions. (<b>A</b>) <span class="html-italic">AtCBF1</span>, (<b>B</b>) <span class="html-italic">AtCBF3</span>, (<b>C</b>) <span class="html-italic">AtCOR15a</span>, (<b>D</b>) <span class="html-italic">AtRD29a</span> relative expression levels. There is an error bar (<span class="html-italic">n</span> = 3) that shows the SD. ** <span class="html-italic">p</span>-value ≤ 0.01.</p>
Full article ">Figure 10
<p>A potential mechanistic model of PfMYB90 adaptation to cold and salt stress.</p>
Full article ">
17 pages, 3794 KiB  
Article
Exploring the Impact of Pharmaceutical Excipient PEG400 on the Pharmacokinetics of Mycophenolic Acid Through In Vitro and In Vivo Experiments
by Chaoji Li, Min Zhang, Yanni Zhao, Dan Yang, Mei Zhao, Leyuan Shang, Xiaodong Sun, Shuo Zhang, Pengjiao Wang and Xiuli Gao
Int. J. Mol. Sci. 2025, 26(1), 72; https://doi.org/10.3390/ijms26010072 - 25 Dec 2024
Viewed by 463
Abstract
Mycophenolic acid (MPA) is a commonly used immunosuppressant. In the human body, MPA is metabolized into mycophenolic acid 7-O-glucuronide (MPAG) and mycophenolic acid acyl-glucuronide (AcMPAG) mainly through liver glucuronidation, which involves UDP-glucuronosyltransferase (UGTs) and transfer proteins. Research has indicated that the pharmaceutical excipient [...] Read more.
Mycophenolic acid (MPA) is a commonly used immunosuppressant. In the human body, MPA is metabolized into mycophenolic acid 7-O-glucuronide (MPAG) and mycophenolic acid acyl-glucuronide (AcMPAG) mainly through liver glucuronidation, which involves UDP-glucuronosyltransferase (UGTs) and transfer proteins. Research has indicated that the pharmaceutical excipient PEG400 can impact drug processes in the body, potentially affecting the pharmacokinetics of MPA. Due to the narrow therapeutic window of MPA, combination therapy is often used, and PEG400 is widely used in pharmaceutical preparations. Therefore, investigating the pharmacokinetic influence of PEG400 on MPA could offer valuable insights for optimizing MPA’s clinical use. In this study, we examined the impact of a single oral dose of PEG400 on the blood levels of MPA in rats through pharmacokinetic analysis. We also investigated the distribution of MPA in various tissues using mass spectrometry imaging. We explored the potential mechanism by which PEG400 affects the metabolism of MPA using hepatic and intestinal microsomes and the Caco-2 cellular transporter model. Our findings reveal that the overall plasma concentrations of MPA were elevated in rats following the co-administration of PEG400, with the AUC0-t of MPA and its metabolite MPAG increasing by 45.53% and 29.44%, respectively. Mass spectrometry imaging showed increased MPA content in tissues after PEG400 administration, with significant differences in the metabolites observed across different tissues. Microsomal and transport experiments showed that PEG400 accelerated the metabolism of MPA, promoted the uptake of MPA, and inhibited efflux. In conclusion, PEG400 alters the in vivo metabolism of MPA, potentially through the modulation of metabolic enzymes and transport. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Structural diagram of the metabolism of MMF (mycophenolate mofetil), MPA (mycophenolic acid), MPAG (mycophenolic acid 7-O-glucuronide), and AcMPAG (mycophenolic acid acyl glucuronide).</p>
Full article ">Figure 2
<p>Comparison of MPA blood concentrations in the presence and absence of PEG400. (<b>A</b>) Blood concentration–time curve of MPA, (<b>B</b>) blood concentration–time curve of MPAG, (<b>C</b>) AUC of MPA and MPAG. * <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; **** <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">n</span> = 6.</p>
Full article ">Figure 3
<p>Spatial mass spectrometry imaging results. (<b>A</b>) Distribution and intensity results of MPA in the liver, kidney, heart, and spleen. (<b>B</b>) Distribution and intensity results of metabolites of MPA in the liver, kidney, heart, and spleen (In the liver, kidney and heart the control group on the left and the PEG400 group on the right, in the spleen the control group on the upper side and the PEG400 group on the lower side). * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 4
<p>Relationship between incubation time and MPAG and AcMPAG concentrations. (<b>A</b>) MPAG concentration over time; (<b>B</b>) AcMPAG concentration over time.</p>
Full article ">Figure 5
<p>Effects of PEG400 on metabolic enzymes (UGTs) in microsomes. Changes in (<b>A</b>) MPAG and (<b>B</b>) AcMPAG concentrations after incubation of MPA (10, 20, 40 μM) with different concentrations of PEG400 (0, 2, 4, 8 μM) in liver microsomes. Changes in (<b>C</b>) MPAG and (<b>D</b>) AcMPAG concentrations after incubation of MPA (10, 20, 40 μM) with different concentrations of PEG400 (0, 2, 4, 8 μM) in intestinal microsomes. * <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; **** <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 6
<p>Cell viability. (<b>A</b>) Effects of different concentrations of MPA on Caco-2 cell survival; (<b>B</b>) Effects of different concentrations of PEG400 on Caco-2 cell survival. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001. Means ± standard deviations, <span class="html-italic">n</span> = 5.</p>
Full article ">Figure 7
<p>Results of MPA transport by PEG400 in the Caco-2 monolayer epithelial cell model. (<b>A</b>) Apparent permeability coefficient Papp values of MPA (40 µM) transport in the Caco-2 cell monolayer model. (<b>B</b>) Effects of PEG400 on ER of MPA (40 µM) in Caco-2 cells. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Means ± standard deviations, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 8
<p>Experimental mechanism diagram. Following oral administration of MMF, MMF is hydrolysed in the gastrointestinal tract to MPA, which is then absorbed into the bloodstream in the intestines.MPA passes through the circulation to the liver, where it is metabolised by the enzyme UGT to MPAG and AcMPAG.A significant amount of MPAG is excreted into the bile, where it is converted by intestinal flora to MPA and reabsorbed into the bloodstream.PEG400 transport affects UGT and transporter proteins, thereby altering the pharmacokinetics of MPA. The transport of PEG400 affects UGT and transporter proteins, thereby altering the pharmacokinetics of MPA.</p>
Full article ">
13 pages, 2170 KiB  
Article
The Difference a Year Can Make: How Antibiotic Resistance Mechanisms in Pseudomonas aeruginosa Have Changed in Northwestern Transylvania
by Matei-Ștefan Dobrescu, Dan-Alexandru Țoc, Adrian-Gabriel Pană, Carmen Costache and Anca Butiuc-Keul
Biomolecules 2025, 15(1), 1; https://doi.org/10.3390/biom15010001 - 24 Dec 2024
Viewed by 574
Abstract
This study examines the prevalence and the mechanisms of antibiotic resistance in Pseudomonas aeruginosa isolates collected from healthcare units in Northwestern Transylvania, Romania, between 2022 and 2023. Given the alarming rise in antibiotic resistance, the study screened 34 isolates for resistance to 10 [...] Read more.
This study examines the prevalence and the mechanisms of antibiotic resistance in Pseudomonas aeruginosa isolates collected from healthcare units in Northwestern Transylvania, Romania, between 2022 and 2023. Given the alarming rise in antibiotic resistance, the study screened 34 isolates for resistance to 10 antibiotics, 46 ARGs, and integrase genes using PCR analysis. The results reveal a concerning increase in multidrug-resistant (MDR) and extensively drug-resistant (XDR) isolates over the two-year period. Notably, the prevalence of ARGs encoding resistance to sulfonamides and beta-lactams, particularly sul1 and blaOXA-50, has shown a significant rise. Furthermore, the study detected the emergence of new resistance mechanisms in the same time interval. These include target protection and even more specific mechanisms, such as metallo-beta-lactamases or enzymes involved in the methylation of 23S rRNA. Statistical analysis further confirmed the correlation between Class I integrons and several ARGs, underscoring the role of horizontal gene transfer in the dissemination of resistance. These findings emphasize the urgent need for updated treatment strategies and monitoring programs to effectively combat the spread of ARGs in clinical settings. Full article
(This article belongs to the Special Issue Novel Mechanisms of Bacterial Antibiotic Resistance)
Show Figures

Figure 1

Figure 1
<p>Workflow of the analysis of the antibiotic resistance of <span class="html-italic">P. aeruginosa</span> isolates.</p>
Full article ">Figure 2
<p>Prevalence of commonly identified ARGs in <span class="html-italic">P. aeruginosa</span> isolates between 2022 and 2023.</p>
Full article ">Figure 3
<p>Prevalence of ARGs in P. aeruginosa isolates by year.</p>
Full article ">Figure 4
<p>Prevalence of genotypic resistance to commonly identified classes of antibiotics in <span class="html-italic">P. aeruginosa</span> isolates between 2022 and 2023.</p>
Full article ">Figure 5
<p>Prevalence of MDR <span class="html-italic">P. aeruginosa</span> isolates between 2022 (<b>a</b>) and 2023 (<b>b</b>).</p>
Full article ">Figure 6
<p>Prevalence of integrons in <span class="html-italic">P. aeruginosa</span> isolates between 2022 and 2023.</p>
Full article ">
Back to TopTop